Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Physiol ; 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38687681

RESUMEN

Altered autonomic input to the heart plays a major role in atrial fibrillation (AF). Autonomic neurons termed ganglionated plexi (GP) are clustered on the heart surface to provide the last point of neural control of cardiac function. To date the properties of GP neurons in humans are unknown. Here we have addressed this knowledge gap in human GP neuron structure and physiology in patients with and without AF. Human right atrial GP neurons embedded in epicardial adipose tissue were excised during open heart surgery performed on both non-AF and AF patients and then characterised physiologically by whole cell patch clamp techniques. Structural analysis was also performed after fixation at both the single cell and at the entire GP levels via three-dimensional confocal imaging. Human GP neurons were found to exhibit unique properties and structural complexity with branched neurite outgrowth. Significant differences in excitability were revealed between AF and non-AF GP neurons as measured by lower current to induce action potential firing, a reduced occurrence of low action potential firing rates, decreased accommodation and increased synaptic density. Visualisation of entire GPs showed almost all neurons are cholinergic with a small proportion of noradrenergic and dual phenotype neurons. Phenotypic distribution differences occurred with AF including decreased cholinergic and dual phenotype neurons, and increased noradrenergic neurons. These data show both functional and structural differences occur between GP neurons from patients with and without AF, highlighting that cellular plasticity occurs in neural input to the heart that could alter autonomic influence on atrial function. KEY POINTS: The autonomic nervous system plays a critical role in regulating heart rhythm and the initiation of AF; however, the structural and functional properties of human autonomic neurons in the autonomic ganglionated plexi (GP) remain unknown. Here we perform the first whole cell patch clamp electrophysiological and large tissue confocal imaging analysis of these neurons from patients with and without AF. Our data show human GP neurons are functionally and structurally complex. Measurements of action potential kinetics show higher excitability in GP neurons from AF patients as measured by lower current to induce action potential firing, reduced low firing action potential rates, and decreased action potential accommodation. Confocal imaging shows increased synaptic density and noradrenergic phenotypes in patients with AF. Both functional and structural differences occur in GP neurons from patients with AF that could alter autonomic influence on atrial rhythm.

2.
Biology (Basel) ; 13(2)2024 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-38392323

RESUMEN

The cardiac autonomic nervous system (CANS) plays a pivotal role in cardiac homeostasis as well as in cardiac pathology. The first level of cardiac autonomic control, the intrinsic cardiac nervous system (ICNS), is located within the epicardial fat pads and is physically organized in ganglionated plexi (GPs). The ICNS system does not only contain parasympathetic cardiac efferent neurons, as long believed, but also afferent neurons and local circuit neurons. Thanks to its high degree of connectivity, combined with neuronal plasticity and memory capacity, the ICNS allows for a beat-to-beat control of all cardiac functions and responses as well as integration with extracardiac and higher centers for longer-term cardiovascular reflexes. The present review provides a detailed overview of the current knowledge of the bidirectional connection between the ICNS and the most studied cardiac pathologies/conditions (myocardial infarction, heart failure, arrhythmias and heart transplant) and the potential therapeutic implications. Indeed, GP modulation with efferent activity inhibition, differently achieved, has been studied for atrial fibrillation and functional bradyarrhythmias, while GP modulation with efferent activity stimulation has been evaluated for myocardial infarction, heart failure and ventricular arrhythmias. Electrical therapy has the unique potential to allow for both kinds of ICNS modulation while preserving the anatomical integrity of the system.

3.
AIChE J ; 69(4)2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37250861

RESUMEN

The baroreflex is a multi-input, multi-output control physiological system that regulates blood pressure by modulating nerve activity between the brainstem and the heart. Existing computational models of the baroreflex do not explictly incorporate the intrinsic cardiac nervous system (ICN), which mediates central control of the heart function. We developed a computational model of closed-loop cardiovascular control by integrating a network representation of the ICN within central control reflex circuits. We examined central and local contributions to the control of heart rate, ventricular functions, and respiratory sinus arrhythmia (RSA). Our simulations match the experimentally observed relationship between RSA and lung tidal volume. Our simulations predicted the relative contributions of the sensory and the motor neuron pathways to the experimentally observed changes in the heart rate. Our closed-loop cardiovascular control model is primed for evaluating bioelectronic interventions to treat heart failure and renormalize cardiovascular physiology.

4.
Front Synaptic Neurosci ; 15: 1104736, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37082542

RESUMEN

The intrinsic cardiac nervous system (ICNS) is composed of interconnected clusters of neurons called ganglionated plexi (GP) which play a major role in controlling heart rate and rhythm. The function of these neurons is particularly important due to their involvement in cardiac arrhythmias such as atrial fibrillation (AF), and previous work has shown that plasticity in GP neural networks could underpin aberrant activity patterns that drive AF. As research in this field increases, developing new techniques to visualize the complex interactions and plasticity in this GP network is essential. In this study we have developed a calcium imaging method enabling the simultaneous recording of plasticity in neuronal activity from multiple neurons in intact atrial GP networks. Calcium imaging was performed with Cal-520 AM labeling in aged spontaneously hypertensive rats (SHRs), which display both spontaneous and induced AF, and age-matched Wistar Kyoto (WKY) controls to determine the relationship between chronic hypertension, arrhythmia and GP calcium dynamics. Our data show that SHR GPs have significantly larger calcium responses to cholinergic stimulation compared to WKY controls, as determined by both higher amplitude and longer duration calcium responses. Responses were significantly but not fully blocked by hexamethonium, indicating multiple cholinergic receptor subtypes are involved in the calcium response. Given that SHRs are susceptible to cardiac arrhythmias, our data provide evidence for a potential link between arrhythmia and plasticity in calcium dynamics that occur not only in cardiomyocytes but also in the GP neurons of the heart.

5.
JACC Clin Electrophysiol ; 9(3): 371-384, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36752452

RESUMEN

BACKGROUND: The sympathetic nervous system plays an integral role in cardiac physiology. Nerve fibers innervating the left ventricle are amenable to transvenous catheter stimulation along the coronary sinus (CS). OBJECTIVES: The aim of the present study was to modulate left ventricular control by selective intracardiac sympathetic denervation. METHODS: First, the impact of epicardial CS ablation on cardiac electrophysiology was studied in a Langendorff model of decentralized murine hearts (n = 10 each, ablation and control groups). Second, the impact of transvenous, anatomically driven axotomy by catheter-based radiofrequency ablation via the CS was evaluated in healthy sheep (n = 8) before and during stellate ganglion stimulation. RESULTS: CS ablation prolonged epicardial ventricular refractory period without (41.8 ± 8.4 ms vs 53.0 ± 13.5 ms; P = 0.049) and with ß1-2-adrenergic receptor blockade (47.8 ± 7.8 ms vs 73.1 ± 13.2 ms; P < 0.001) in mice. Supported by neuromorphological studies illustrating a circumferential CS neural network, intracardiac axotomy by catheter ablation via the CS in healthy sheep diminished the blood pressure increase during stellate ganglion stimulation (Δ systolic blood pressure 21.9 ± 10.9 mm Hg vs 10.5 ± 12.0 mm Hg; P = 0.023; Δ diastolic blood pressure 9.0 ± 5.5 mm Hg vs 3.0 ± 3.5 mm Hg; P = 0.039). CONCLUSIONS: Transvenous, anatomically driven axotomy targeting nerve fibers along the CS enables acute modulation of left ventricular control by selective intracardiac sympathetic denervation.


Asunto(s)
Ventrículos Cardíacos , Corazón , Animales , Ratones , Ovinos , Ventrículos Cardíacos/cirugía , Ventrículos Cardíacos/inervación , Simpatectomía , Sistema Nervioso Simpático/cirugía , Sistema Nervioso Simpático/fisiología , Ganglio Estrellado/cirugía
6.
J Comp Neurol ; 531(5): 596-617, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36591925

RESUMEN

Sympathetic efferent axons regulate cardiac functions. However, the topographical distribution and morphology of cardiac sympathetic efferent axons remain insufficiently characterized due to the technical challenges involved in immunohistochemical labeling of the thick walls of the whole heart. In this study, flat-mounts of the left and right atria and ventricles of FVB mice were immunolabeled for tyrosine hydroxylase (TH), a marker of sympathetic nerves. Atrial and ventricular flat-mounts were scanned using a confocal microscope to construct montages. We found (1) In the atria: A few large TH-immunoreactive (IR) axon bundles entered both atria, branched into small bundles and then single axons that eventually formed very dense terminal networks in the epicardium, myocardium and inlet regions of great vessels to the atria. Varicose TH-IR axons formed close contact with cardiomyocytes, vessels, and adipocytes. Multiple intrinsic cardiac ganglia (ICG) were identified in the epicardium of both atria, and a subpopulation of the neurons in the ICG were TH-IR. Most TH-IR axons in bundles traveled through ICG before forming dense varicose terminal networks in cardiomyocytes. We did not observe varicose TH-IR terminals encircling ICG neurons. (2) In the left and right ventricles and interventricular septum: TH-IR axons formed dense terminal networks in the epicardium, myocardium, and vasculature. Collectively, TH labeling is achievable in flat-mounts of thick cardiac walls, enabling detailed mapping of catecholaminergic axons and terminal structures in the whole heart at single-cell/axon/varicosity scale. This approach provides a foundation for future quantification of the topographical organization of the cardiac sympathetic innervation in different pathological conditions.


Asunto(s)
Ventrículos Cardíacos , Corazón , Ratones , Animales , Ventrículos Cardíacos/inervación , Inmunohistoquímica , Corazón/inervación , Axones , Miocardio , Tirosina 3-Monooxigenasa
7.
Europace ; 25(2): 726-738, 2023 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-36260428

RESUMEN

AIMS: The response to high frequency stimulation (HFS) is used to locate putative sites of ganglionated plexuses (GPs), which are implicated in triggering atrial fibrillation (AF). To identify topological and immunohistochemical characteristics of presumed GP sites functionally identified by HFS. METHODS AND RESULTS: Sixty-three atrial sites were tested with HFS in four Langendorff-perfused porcine hearts. A 3.5 mm tip quadripolar ablation catheter was used to stimulate and deliver HFS to the left and right atrial epicardium, within the local atrial refractory period. Tissue samples from sites triggering atrial ectopy/AF (ET) sites and non-ET sites were stained with choline acetyltransferase (ChAT) and tyrosine hydroxylase (TH), for quantification of parasympathetic and sympathetic nerves, respectively. The average cross-sectional area (CSA) of nerves was also calculated. Histomorphometry of six ET sites (9.5%) identified by HFS evoking at least a single atrial ectopic was compared with non-ET sites. All ET sites contained ChAT-immunoreactive (ChAT-IR) and/or TH-immunoreactive nerves (TH-IR). Nerve density was greater in ET sites compared to non-ET sites (nerves/cm2: 162.3 ± 110.9 vs. 69.65 ± 72.48; P = 0.047). Overall, TH-IR nerves had a larger CSA than ChAT-IR nerves (µm2: 11 196 ± 35 141 vs. 2070 ± 5841; P < 0.0001), but in ET sites, TH-IR nerves were smaller than in non-ET sites (µm2: 6021 ± 14 586 vs. 25 254 ± 61 499; P < 0.001). CONCLUSIONS: ET sites identified by HFS contained a higher density of smaller nerves than non-ET sites. The majority of these nerves were within the atrial myocardium. This has important clinical implications for devising an effective therapeutic strategy for targeting autonomic triggers of AF.


Asunto(s)
Fibrilación Atrial , Ablación por Catéter , Animales , Porcinos , Fibrilación Atrial/cirugía , Atrios Cardíacos , Miocardio , Sistema Nervioso Autónomo , Ablación por Catéter/métodos
8.
Front Physiol ; 14: 1284673, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38179138

RESUMEN

Aims: The behavior of pacemaker cardiomyocytes (PCs) in the sinoatrial node (SAN) is modulated by neurohormonal and paracrine factors, many of which signal through G-protein coupled receptors (GPCRs). The aims of the present study are to catalog GPCRs that are differentially expressed in the mammalian SAN and to define the acute physiological consequences of activating the cholecystokinin-A signaling system in isolated PCs. Methods and results: Using bulk and single cell RNA sequencing datasets, we identify a set of GPCRs that are differentially expressed between SAN and right atrial tissue, including several whose roles in PCs and in the SAN have not been thoroughly characterized. Focusing on one such GPCR, Cholecystokinin-A receptor (CCKAR), we demonstrate expression of Cckar mRNA specifically in mouse PCs, and further demonstrate that subsets of SAN fibroblasts and neurons within the cardiac intrinsic nervous system express cholecystokinin, the ligand for CCKAR. Using mouse models, we find that while baseline SAN function is not dramatically affected by loss of CCKAR, the firing rate of individual PCs is slowed by exposure to sulfated cholecystokinin-8 (sCCK-8), the high affinity ligand for CCKAR. The effect of sCCK-8 on firing rate is mediated by reduction in the rate of spontaneous phase 4 depolarization of PCs and is mitigated by activation of beta-adrenergic signaling. Conclusion: (1) PCs express many GPCRs whose specific roles in SAN function have not been characterized, (2) Activation of the cholecystokinin-A signaling pathway regulates PC automaticity.

9.
Elife ; 112022 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-36426848

RESUMEN

Stellate ganglia within the intrathoracic cardiac control system receive and integrate central, peripheral, and cardiopulmonary information to produce postganglionic cardiac sympathetic inputs. Pathological anatomical and structural remodeling occurs within the neurons of the stellate ganglion (SG) in the setting of heart failure (HF). A large proportion of SG neurons function as interneurons whose networking capabilities are largely unknown. Current therapies are limited to targeting sympathetic activity at the cardiac level or surgical interventions such as stellectomy, to treat HF. Future therapies that target the SG will require understanding of their networking capabilities to modify any pathological remodeling. We observe SG networking by examining cofluctuation and specificity of SG networked activity to cardiac cycle phases. We investigate network processing of cardiopulmonary transduction by SG neuronal populations in porcine with chronic pacing-induced HF and control subjects during extended in-vivo extracellular microelectrode recordings. We find that information processing and cardiac control in chronic HF by the SG, relative to controls, exhibits: (i) more frequent, short-lived, high magnitude cofluctuations, (ii) greater variation in neural specificity to cardiac cycles, and (iii) neural network activity and cardiac control linkage that depends on disease state and cofluctuation magnitude.


Asunto(s)
Insuficiencia Cardíaca , Ganglio Estrellado , Animales , Porcinos , Ganglio Estrellado/fisiología , Ganglio Estrellado/cirugía , Benchmarking , Entropía , Corazón
10.
Front Physiol ; 13: 835761, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35574437

RESUMEN

Neural control of the heart involves continuous modulation of cardiac mechanical and electrical activity to meet the organism's demand for blood flow. The closed-loop control scheme consists of interconnected neural networks with central and peripheral components working cooperatively with each other. These components have evolved to cooperate control of various aspects of cardiac function, which produce measurable "functional" outputs such as heart rate and blood pressure. In this review, we will outline fundamental studies probing the cardiac neural control hierarchy. We will discuss how computational methods can guide improved experimental design and be used to probe how information is processed while closed-loop control is operational. These experimental designs generate large cardio-neural datasets that require sophisticated strategies for signal processing and time series analysis, while presenting the usual large-scale computational challenges surrounding data sharing and reproducibility. These challenges provide unique opportunities for the development and validation of novel techniques to enhance understanding of mechanisms of cardiac pathologies required for clinical implementation.

11.
Ann Anat ; 242: 151911, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35183709

RESUMEN

BACKGROUND: No reports have been made on the entire extrinsic innervation of the heart in small laboratory animals. Therefore, this study examined the detailed morphotopographic features of the extrinsic cardiac autonomic nervous system (ECANS) with its adjacent structures (1) to record the general morpho-topography and variations of the ECANS in guinea pigs, (2) to compare it with previous reports on common laboratory rodents (rats, mice, and Syrian hamsters), rabbits, domesticated animals (cats, dogs, sheep, goats, oxen, pigs, and horses), primates, and humans, and (3) to infer the macroscopic evolutionary changes they presented. METHODS: The sympathetic ganglia, vagi, and emitting cardiac nerves/branches in the cervical and thoracic regions were dissected in 24 sides of 12 formalin-fixed, arterially injected adult male and female guinea pigs under a stereomicroscope. RESULTS: The ECANS in guinea pigs presented following general morphologic characteristics: (1) constant existence of the cranial cervical ganglion (CG) and placing caudal to the cranial base over the ventrolateral aspect of the longus capitis muscle, dorsomedial to the common carotid artery and communicating to the first two cervical spinal nerves, (2) the lack of the vago-sympathetic trunk, (3) the existence of the middle cervical ganglion (MG) and lying on the lateral aspect of the longus colli muscle (LC) at the level of the seventh cervical vertebra, (4) constant existence of the cervicothoracic ganglion (CT) composing generally from the caudal cervical ganglion and 1-3 thoracic ganglia and placing ventral to the first and second intercostal spaces over the lateral aspect of the LC and communicating to the eight cervical and first three thoracic spinal nerves in addition to the vertebral nerve, (5) constant existence of the limbs of the ansa subclavia (AS) joining the CT to MG, (6) the existence of individual thoracic ganglia from the 4th to the 12th and joining by single interganglionic branches (IGBs), and communicating to corresponding thoracic nerve, (7) the intimate relation between the caudal part of the thoracic sympathetic chain and the quadratus lumborum muscle, (8) the main cardiac nerves (CNs) emerging from the CT, (9) the lack of CNs springing generally from the CG, ST, AS, MG, or individual thoracic ganglia or their IGBs, and (10) the existence of the cardiac branches (CBs) emerging from the vagi and recurrent laryngeal nerves. The ECANS morphology in guinea pigs also shows sex and laterality differences. CONCLUSIONS: The general anatomical arrangement of the sympathetic components of the ECANS in guinea pigs extremely displaced features common to rats and Syrian hamsters regardless of the existence of MG and the close relation between the thoracic sympathetic chain and the quadratus lumborum muscle. However, the position and organization of the CT, along with its rami communicantes to spinal nerves in guinea pigs quite resembled those seen in rats. The general macroscopic arrangement of the sympathetic components of the ECANS in guinea pigs resembled that seen in rabbits regardless of the organization and location of the CT. The general morphology of the sympathetic components of the ECANS demonstrated markedly morphological variations and similarities among common laboratory rodents, rabbits, domesticated animals (DNs), primates, and humans. The main variations consisted of the position of the CG and its rami communicantes with the spinal nerves, the relation between the vagi and sympathetic trunks in the neck, the existence of the MG, the location and arrangement of the CT, the origins and incidences of the cardiac nerves, and the main sympathetic contributors. The general macroscopic architecture of the parasympathetic components of the ECANS in guinea pigs quite resembled that seen in domesticated animals, primates, and humans. Evolutionary comparative morphologic characteristics of the ECANS are discussed in detail and evolutionary differences and similarities of the ECANS have been found from common laboratory rodents, rabbits, domesticated animals, and primates to humans.


Asunto(s)
Ganglios Simpáticos , Cobayas , Sistema Nervioso Simpático , Animales , Sistema Nervioso Autónomo/anatomía & histología , Femenino , Ganglios Simpáticos/anatomía & histología , Cobayas/anatomía & histología , Corazón , Humanos , Masculino , Mamíferos , Cuello , Sistema Nervioso Simpático/anatomía & histología
12.
Ann Anat ; 239: 151842, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34673201

RESUMEN

BACKGROUND: The detailed morphology and topography of the sympathetic cardiac nerves (SCNs) and ganglia with their surrounding structures in Syrian hamsters were examined to document the general topography and morphology and variations and to discuss the comparative anatomy between the SCNs and ganglia in Syrian hamsters and other rodents, as well as their comparative morphology and macroscopic evolutionary changes among rodents, rabbits, domestic animals (cats, dogs, sheep, goats, oxen, pigs and horses), primates, and humans. METHODS: The composition of the cervical and thoracic parts of the sympathetic trunks and ganglia was bilaterally microdissected in twenty-eight sides of 14 adult male and female Syrian hamsters under a stereomicroscope. RESULTS: The general morphology of the SCNs and related ganglia in Syrian hamsters was obtained and noted as follows: (1) the absence of the vago-sympathetic trunk, (2) the absence of the middle cervical ganglion (MG), (3) constant presence of the cervicothoracic ganglion (CT) comprising generally from the caudal cervical ganglion and 1-2 thoracic ganglia and locating over the lateral surface of the longus colli muscle ventral to the heads of the first two ribs and communicating to the eight cervical and first two thoracic spinal nerves (C8-T2) in addition to the vertebral nerve, (4) extensive coverage of the lateral surface of the CT by branches of the subclavian artery, (5) the cranial and caudal limbs of the ansa subclavia (AS) joining the CT to the caudal end of the cervical sympathetic trunk, (6) the presence of an independent thoracic ganglion from the 2nd or 3rd to the 13th and connecting by single interganglionic branches, and communicating to each thoracic spinal nerve, (7) close relationship between the caudal portion of the thoracic sympathetic trunk and the psoas minor muscle, (8) the primary cardiac nerves (CNs) arising from the CT, and (9) the absence of CNs originating generally from the cervical sympathetic trunk, AS, MG, or independent thoracic ganglia or their interganglionic branches. Individual variations of the SCNs and ganglia in Syrian hamsters were noted, including the absence of the ansa subclavia on 5/28 sides (17.86%), the presence of the intermediate ganglia (IG) placed on the C7 on 3/28 sides (10.71%) or the C8 on 3/28 sides (10.71%), and no CNs arising from the IG as well as the presence of the double thoracic sympathetic trunk on 5/28 sides (17.86%). The anatomical characteristics of the SCNs and related ganglia were also exhibited sex and laterality differences. CONCLUSIONS: From a comparative anatomy viewpoint, the general morphology of the SCNs and related ganglia in Syrian hamsters was very similar to that in rats but was considerably different from that in guinea pigs, especially concerning the MG, cranial position and composition of the CT. The general morphology of the SCNs and related ganglia in Syrian hamsters and other laboratory rodents resembled that of rabbits but was essentially different from that in rabbits with respect to the cranial position and composition of the CT. The general morphology of the SCNs and ganglia exhibited significant morphological differences and similarities among laboratory rodents, rabbits, domestic animals, primates, and humans. The main differences include the relationship between the cervical parts of the vagus nerve and sympathetic trunk, the presence of the MG, the position and composition of the CT, the origins and frequencies of the cardiac nerves, and the primary sympathetic contributor. From macroscopic evolutionary change, the expansion of the range of the SCNs origin has occurred from laboratory rodents, rabbits, domestic animals, and primates to humans.


Asunto(s)
Ganglios Simpáticos , Sistema Nervioso Simpático , Animales , Cricetinae , Femenino , Cobayas , Caballos , Masculino , Mesocricetus , Cuello , Conejos , Ratas , Ovinos , Tórax
14.
Exp Gerontol ; 148: 111261, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33647361

RESUMEN

Hypertriglyceridemia is a result of the increase in the serum levels of lipoproteins, which are responsible for the transport of triglycerides and can be caused by genetic and/or metabolic factors. Animal models which either express or lack genes related to changes in the lipoproteins profile are useful to understand lipid metabolism. Apolipoprotein CIII (apoCIII) is an important modulator of hepatic production and peripheral removal of triglycerides. Mice that overexpress the apoCIII gene become hypertriglyceridemic, showing high concentrations of free fatty acids in the blood. Since hypertriglyceridemia is related to atherosclerosis, and the latter refers to cardiac alterations, this study aimed at evaluating the morphological, morphometric and quantitative profiles of the cardiac plexus, as well as the morphometric and histopathological aspects of the epicardial adipose tissue in human apoCIII transgenic mice. Therefore, 8-12-month-old male C57BL/6 mice that overexpressed human apoCIII (CIII) and their respective controls were used. Our results showed that overexpression of human apoCIII did not modify morphological or quantitative parameters of cardiac plexus neurons; however, age increased both, the area and the number of such cells. Furthermore, there was a direct correlation of this dyslipidemia to the thickening of periganglionar type 1 collagens. On the other hand, this overexpression caused epicardial adipose tissue inflammation and an increase in the area of the adipocytes, thus, favoring the recruitment of inflammatory cells in this tissue. In conclusion, this overexpression is harmful since it is related to an increase in cardiac adiposity, as well as to a predisposition to an inflammatory environment in the epicardial fat and to the incidence of cardiovascular diseases.


Asunto(s)
Tejido Adiposo , Inflamación , Animales , Apolipoproteína C-III , Humanos , Inflamación/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Triglicéridos
15.
Heart Rhythm O2 ; 1(3): 215-221, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34113874

RESUMEN

BACKGROUND: Recent animal and human studies have shown antiarrhythmic effects inhibiting inducibility of atrial fibrillation through low-level transcutaneous electrical stimulation at the auricular branch of the vagus nerve (ABVN). OBJECTIVE: The present study investigated effects of acupuncture at the ABVN on the autonomic cardiac nervous system in humans through analysis of heart rate and heart rate variability (HRV) parameters. METHODS: We enrolled 24 healthy male volunteers and compared acupuncture at the ABVN to placebo-acupuncture performed at the Ma-35 point (an acupuncture point used in traditional Chinese medicine to treat pain caused by gonarthrosis). An additional measurement without acupuncture served as control. We analyzed the following heart rate and HRV parameters: standard deviation of normal-to-normal intervals (SDNN), root mean square of successive R-R interval differences (RMSSD), high frequency (HF), low frequency (LF), LF/HF ratio. RESULTS: In comparison to placebo acupuncture, acupuncture at the ABVN led to a significant reduction in heart rate (approximately 4%-6%, P < .05) and an increase in overall HRV demonstrated by SDNN (approximately 19%, P < .05). RMSSD and power spectral density parameters (HF, LF, LF/HF) showed statistical trends (P < .1) induced by auricular acupuncture in favor of vagal tone. No relevant difference was shown between control and placebo group. CONCLUSION: Acupuncture of the region innervated by the ABVN may activate the parasympathetic nervous system, as suggested by reduction in heart rate and increase in SDNN. However, given the lack of clear significant changes in other HRV parameters, this effect seems modest and its evaluation requires further investigation.

16.
Med Hypotheses ; 129: 109253, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31371087

RESUMEN

Ivabradine decreases heart rate by selective inhibition of the If current in the sinoatrial node. Ivabradine is declared to have no direct effect on the autonomic nervous system (ANS). However, there are some data suggesting an (at least indirect) effect of ivabradine on the ANS. The pathomechanism behind is unclear. Based on the complex of plexuses and ganglia in the heart, the existence of the intrinsic cardiac nervous system (ICNS), also known as the "little brain" of the heart, has been suggested. The ICNS is supposed to process information on the cardiac milieu and provide the central nervous system with these data. We put forward a hypothesis that part of ivabradine's protective effects might reside in the modulation of the ANS by affecting the ICNS. Setting a new autonomic balance by ivabradine might be of benefit in the treatment of autonomic dysfunction-related pathologies.


Asunto(s)
Sistema Nervioso Autónomo/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Ivabradina/farmacología , Nodo Sinoatrial/efectos de los fármacos , Animales , Enfermedades del Sistema Nervioso Autónomo/tratamiento farmacológico , Fármacos Cardiovasculares , Insuficiencia Cardíaca/tratamiento farmacológico , Humanos , Modelos Teóricos , Ratas
17.
Card Fail Rev ; 4(2): 92-98, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30206483

RESUMEN

Heart failure (HF) is associated with significant morbidity and mortality. The disease is characterised by autonomic imbalance with increased sympathetic activity and withdrawal of parasympathetic activity. Despite the use of medical therapies that target, in part, the neurohormonal axis, rates of HF progression, morbidity and mortality remain high. Emerging therapies centred on neuromodulation of autonomic control of the heart provide an alternative device-based approach to restoring sympathovagal balance. Preclinical studies have proven favourable, while clinical trials have had mixed results. This article highlights the importance of understanding structural/functional organisation of the cardiac nervous system as mechanistic-based neuromodulation therapies evolve.

18.
Auton Neurosci ; 208: 57-65, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28919363

RESUMEN

BACKGROUND: Autonomic regulation therapy involving either vagus nerve stimulation (VNS) or spinal cord stimulation (SCS) represents emerging bioelectronic therapies for heart disease. The objective of this study was to determine if VNS and/or SCS modulate primary cardiac afferent sensory transduction of the ischemic myocardium. METHODS: Using extracellular recordings in 19 anesthetized canines, of 88 neurons evaluated, 36 ventricular-related nodose ganglia sensory neurons were identified by their functional activity responses to epicardial touch, chemical activation of their sensory neurites (epicardial veratridine) and great vessel (descending aorta or inferior vena cava) occlusion. Neural responses to 1min left anterior descending (LAD) coronary artery occlusion (CAO) were then evaluated. These interventions were then studied following either: i) SCS [T1-T3 spinal level; 50Hz, 90% motor threshold] or ii) cervical VNS [15-20Hz; 1.2× threshold]. RESULTS: LAD occlusion activated 66% of identified nodose ventricular sensory neurons (0.33±0.08-0.79±0.20Hz; baseline to CAO; p<0.002). Basal activity of cardiac-related nodose neurons was differentially reduced by VNS (0.31±0.11 to 0.05±0.02Hz; p<0.05) as compared to SCS (0.36±0.12 to 0.28±0.14, p=0.59), with their activity response to transient LAD CAO being suppressed by either SCS (0.85±0.39-0.11±0.04Hz; p<0.03) or VNS (0.75±0.27-0.12±0.05Hz; p<0.04). VNS did not alter evoked neural responses of cardiac-related nodose neurons to great vessel occlusion. CONCLUSIONS: Both VNS and SCS obtund ventricular ischemia induced enhancement of nodose afferent neuronal inputs to the medulla.


Asunto(s)
Isquemia Miocárdica/fisiopatología , Ganglio Nudoso/fisiopatología , Células Receptoras Sensoriales/fisiología , Columna Vertebral/fisiopatología , Sistema Nervioso Simpático/fisiopatología , Potenciales de Acción , Animales , Modelos Animales de Enfermedad , Perros , Estimulación Eléctrica , Inmunohistoquímica , Microelectrodos , Isquemia Miocárdica/patología , Ganglio Nudoso/patología , Células Receptoras Sensoriales/patología , Columna Vertebral/patología , Sistema Nervioso Simpático/patología , Vértebras Torácicas
19.
World J Cardiol ; 9(6): 508-520, 2017 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-28706586

RESUMEN

Neural elements of the intrinsic cardiac nervous system transduce sensory inputs from the heart, blood vessels and other organs to ensure adequate cardiac function on a beat-to-beat basis. This inter-organ crosstalk is critical for normal function of the heart and other organs; derangements within the nervous system hierarchy contribute to pathogenesis of organ dysfunction. The role of intact cardiac nerves in development of, as well as protection against, ischemic injury is of current interest since it may involve recruitment of intrinsic cardiac ganglia. For instance, ischemic conditioning, a novel protection strategy against organ injury, and in particular remote conditioning, is likely mediated by activation of neural pathways or by endogenous cytoprotective blood-borne substances that stimulate different signalling pathways. This discovery reinforces the concept that inter-organ communication, and maintenance thereof, is key. As such, greater understanding of mechanisms and elucidation of treatment strategies is imperative to improve clinical outcomes particularly in patients with comorbidities. For instance, autonomic imbalance between sympathetic and parasympathetic nervous system regulation can initiate cardiovascular autonomic neuropathy that compromises cardiac stability and function. Neuromodulation therapies that directly target the intrinsic cardiac nervous system or other elements of the nervous system hierarchy are currently being investigated for treatment of different maladies in animal and human studies.

20.
Basic Res Cardiol ; 112(2): 11, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28091727

RESUMEN

This study aimed to investigate the role of the intrinsic cardiac nervous system in the mechanism of classical myocardial ischaemic preconditioning (IPC). Isolated perfused rat hearts were subjected to 35-min regional ischaemia and 60-min reperfusion. IPC was induced as three cycles of 5-min global ischaemia-reperfusion, and provided significant reduction in infarct size (IS/AAR = 14 ± 2% vs control IS/AAR = 48 ± 3%, p < 0.05). Treatment with the ganglionic antagonist, hexamethonium (50 µM), blocked IPC protection (IS/AAR = 37 ± 7%, p < 0.05 vs IPC). Moreover, the muscarinic antagonist, atropine (100 nM), also abrogated IPC-mediated protection (IS/AAR = 40 ± 3%, p < 0.05 vs IPC). This indicates that intrinsic cardiac ganglia remain intact in the Langendorff preparation and are important in the mechanism of IPC. In a second group of experiments, coronary effluent collected following IPC, from ex vivo perfused rat hearts, provided significant cardioprotection when perfused through a naïve isolated rat heart prior to induction of regional ischaemia-reperfusion injury (IRI) (IS/ARR = 19 ± 2, p < 0.05 vs control effluent). This protection was also abrogated by treating the naïve heart with hexamethonium, indicating the humoral trigger of IPC induces protection via an intrinsic neuronal mechanism (IS/AAR = 46 ± 5%, p < 0.05 vs IPC effluent). In addition, a large release in ACh was observed in coronary effluent was observed following IPC (IPCeff = 0.36 ± 0.03 µM vs C eff = 0.04 ± 0.04 µM, n = 4, p < 0.001). Interestingly, however, IPC effluent was not able to significantly protect isolated cardiomyocytes from simulated ischaemia-reperfusion injury (cell death = 45 ± 6%, p = 0.09 vs control effluent). In conclusion, IPC involves activation of the intrinsic cardiac nervous system, leading to release of ACh in the ventricles and induction of protection via activation of muscarinic receptors.


Asunto(s)
Acetilcolina/metabolismo , Ganglios/metabolismo , Corazón/inervación , Precondicionamiento Isquémico Miocárdico , Daño por Reperfusión Miocárdica/fisiopatología , Animales , Modelos Animales de Enfermedad , Preparación de Corazón Aislado , Masculino , Infarto del Miocardio , Daño por Reperfusión Miocárdica/metabolismo , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA