Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Neurosurg Focus ; 56(5): E11, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38691862

RESUMEN

OBJECTIVE: In the treatment of skull base chordoma (SBC) surgery is considered the mainstay approach, and gross-total resection has an established relationship with progression-free survival (PFS) and overall survival (OS). However, the tumor's location often interferes with attempts at complete resection. In this case, surgery for maximal resection followed by high-dose radiotherapy has been demonstrated to be the standard treatment. In this context, various modalities are available, yet no consensus exists on the most effective. This systematic review and meta-analysis aimed to evaluate the efficacy and safety of different radiotherapy modalities for SBC. METHODS: Following PRISMA guidelines, the authors systematically searched for the treatment of SBC with radiation modalities in the PubMed, Cochrane, Web of Science, and EMBASE databases. Outcomes assessed for each modality were as follows: OS, PFS, local control (LC), and complications. The random-effects model was adopted. A single-proportion analysis with 95% CI was used to measure the effects in single-arm analysis. For the comparative analysis, the OR with 95% CI was used to compare outcome treatment effects. Heterogeneity was assessed using I2 statistics, and statistical significance was defined as p < 0.05. RESULTS: A total of 32 studies comprising 3663 patients, with 2322 patients who were treated with radiotherapeutic modalities, were included. Regarding 5-year OS findings in each modality study, the findings were as follows: in photon fractionated radiotherapy, an estimated rate of 77% (69%-84%, 568 patients); in conventional fractionated radiotherapy, 76% (65%-87%, 517 cases); in proton-based + carbon ion-based radiotherapy, 85% (82%-88%, 622 cases); and in a comparative analysis of proton-based and carbon ion-based therapy, there was an OR of 1.2 (95% CI 0.59-2.43, 306 cases). Regarding the 5-year PFS estimate, the rates were as follows: 35% (26%-45%, 95 cases) for photon fractionated therapy; 35% (25%-45%, 85 cases) for stereotactic radiotherapy; 77% (50%-100%, 180 cases) for proton-based and carbon ion-based radiotherapy; and 74% (45%-100%, 102 cases) for proton-based radiotherapy. Regarding LC in periods of 3 and 5 years after proton- and carbon ion-based therapy, the overall estimated rates were 84% (78%-90%, 326 cases) and 75% (65%-85%, 448 cases), respectively. For proton-based radiotherapy and carbon ion-based therapy, the 5-year LC rates were 76% (67%-86%, 259 cases) and 75% (59%-91%, 189 cases), respectively. CONCLUSIONS: The analysis highlights the finding that particle-based modalities like proton beam radiotherapy and carbon ion radiotherapy are the most effective radiation therapies available for the treatment of SBC. Furthermore, it reinforces the idea that surgery followed by radiotherapy constitutes the standard treatment.


Asunto(s)
Cordoma , Neoplasias de la Base del Cráneo , Humanos , Neoplasias de la Base del Cráneo/radioterapia , Neoplasias de la Base del Cráneo/cirugía , Cordoma/radioterapia , Cordoma/cirugía , Resultado del Tratamiento , Radiocirugia/métodos
2.
Int J Mol Sci ; 25(10)2024 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-38791266

RESUMEN

Sympathetic nervous system (SNS) hyperactivity is mediated by elevated catecholamine (CA) secretion from the adrenal medulla, as well as enhanced norepinephrine (NE) release from peripheral sympathetic nerve terminals. Adrenal CA production from chromaffin cells is tightly regulated by sympatho-inhibitory α2-adrenergic (auto)receptors (ARs), which inhibit both epinephrine (Epi) and NE secretion via coupling to Gi/o proteins. α2-AR function is, in turn, regulated by G protein-coupled receptor (GPCR)-kinases (GRKs), especially GRK2, which phosphorylate and desensitize them, i.e., uncouple them from G proteins. On the other hand, the short-chain free fatty acid (SCFA) receptor (FFAR)-3, also known as GPR41, promotes NE release from sympathetic neurons via the Gi/o-derived free Gßγ-activated phospholipase C (PLC)-ß/Ca2+ signaling pathway. However, whether it exerts a similar effect in adrenal chromaffin cells is not known at present. In the present study, we examined the interplay of the sympatho-inhibitory α2A-AR and the sympatho-stimulatory FFAR3 in the regulation of CA secretion from rat adrenal chromaffin (pheochromocytoma) PC12 cells. We show that FFAR3 promotes CA secretion, similarly to what GRK2-dependent α2A-AR desensitization does. In addition, FFAR3 activation enhances the effect of the physiologic stimulus (acetylcholine) on CA secretion. Importantly, GRK2 blockade to restore α2A-AR function or the ketone body beta-hydroxybutyrate (BHB or 3-hydroxybutyrate), via FFAR3 antagonism, partially suppress CA production, when applied individually. When combined, however, CA secretion from PC12 cells is profoundly suppressed. Finally, propionate-activated FFAR3 induces leptin and adiponectin secretion from PC12 cells, two important adipokines known to be involved in tissue inflammation, and this effect of FFAR3 is fully blocked by the ketone BHB. In conclusion, SCFAs can promote CA and adipokine secretion from adrenal chromaffin cells via FFAR3 activation, but the metabolite/ketone body BHB can effectively inhibit this action.


Asunto(s)
Catecolaminas , Receptores Adrenérgicos alfa 2 , Receptores Acoplados a Proteínas G , Animales , Células PC12 , Ratas , Receptores Acoplados a Proteínas G/metabolismo , Catecolaminas/metabolismo , Receptores Adrenérgicos alfa 2/metabolismo , Adipoquinas/metabolismo , Células Cromafines/metabolismo , Transducción de Señal , Norepinefrina/metabolismo , Norepinefrina/farmacología
3.
J Clin Neurosci ; 123: 47-54, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38531194

RESUMEN

BACKGROUND: Endovascular treatment of intracranial aneurysms (EVTIAs) is increasingly popular due to its minimally invasive nature and high success rate. While general anesthesia (GA) has been the historical preference for EVTIAs, there's growing interest in local anesthesia (LA). However, concerns persist about LA safety for EVTIAs. Therefore, we conducted a systematic review and meta-analysis to assess LA safety for EVTIAs. METHODS: Following PRISMA guidelines, we searched PubMed, Embase, and Web of Science databases. Pooled analysis with 95 % confidence intervals (CI) assessed effects, I2 statistics gauged heterogeneity, and a random-effects model was adopted. Conversion to GA, neurological or procedure-related complications, intraoperative intracranial hemorrhagic complications (IIHC), and mortality were assessed. Subanalyses for ruptured and unruptured cases were performed. RESULTS: The analysis included eleven studies, 2,133 patients, and 2,369 EVTIAs under LA. Conversion to GA rate was 1 % (95 %CI: 0 to 2 %). Neurological or procedure-related complications rate was 13 % (95 % CI: 8 % to 17 %). IIHC analysis revealed a rate of 1 % (95 % CI: 1 % to 2 %). The mortality rate was 0 % (95 %CI: 0 % to 0 %). Subanalyses revealed similar rates in ruptured and unruptured subgroups, except for a slightly high rate of complications and IIHC in the ruptured subgroup. CONCLUSION: Findings indicate that EVTIA under LA is safe, with low conversion and mortality rates, even for ruptured aneurysms. Complications rates, also in IIHC rates, are comparable to those reported for GA, emphasizing LA's comparable safety profile in EVTIAs. Considering these promising outcomes, the decision to opt for the LA approach emerges as meaningful and well-suited for the endovascular treatment of aneurysms. Beyond its safety, LA introduces inherent supplementary advantages, including shortened hospitalization periods, cost-effectiveness, and an expedited patient recovery process.


Asunto(s)
Anestesia Local , Procedimientos Endovasculares , Aneurisma Intracraneal , Humanos , Procedimientos Endovasculares/efectos adversos , Procedimientos Endovasculares/métodos , Aneurisma Intracraneal/cirugía , Anestesia Local/métodos
4.
Vitam Horm ; 124: 393-404, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38408805

RESUMEN

The adrenal cortex is responsible for production of adrenal steroid hormones and is anatomically divided into three distinct zones: zona glomerulosa secreting mineralocorticoids (mainly aldosterone), zona fasciculata secreting glucocorticoids (cortisol), and zona reticularis producing androgens. Importantly, due to their high lipophilicity, no adrenal steroid hormone (including aldosterone) is stored in vesicles but rather gets synthesized and secreted instantly upon cell stimulation with specific stimuli. Aldosterone is the most potent mineralocorticoid hormone produced from the adrenal cortex in response to either angiotensin II (AngII) or elevated K+ levels in the blood (hyperkalemia). AngII, being a peptide, cannot cross cell membranes and thus, uses two distinct G protein-coupled receptor (GPCR) types, AngII type 1 receptor (AT1R) and AT2R to exert its effects inside cells. In zona glomerulosa cells, AT1R activation by AngII results in aldosterone synthesis and secretion via two main pathways: (a) Gq/11 proteins that activate phospholipase C ultimately raising intracellular free calcium concentration; and (b) ßarrestin1 and -2 (also known as Arrestin-2 and -3, respectively) that elicit sustained extracellular signal-regulated kinase (ERK) activation. Both pathways induce upregulation and acute activation of StAR (steroidogenic acute regulatory) protein, the enzyme that catalyzes the rate-limiting step in aldosterone biosynthesis. This chapter describes these two salient pathways underlying AT1R-induced aldosterone production in zona glomerulosa cells. We also highlight some pharmacologically important notions pertaining to the efficacy of the currently available AT1R antagonists, also known as angiotensin receptor blockers (ARBs) or sartans at suppressing both pathways, i.e., their inverse agonism efficacy at G proteins and ßarrestins.


Asunto(s)
Corteza Suprarrenal , Aldosterona , Humanos , Aldosterona/metabolismo , Angiotensina II , Antagonistas de Receptores de Angiotensina/farmacología , Agonismo Inverso de Drogas , Inhibidores de la Enzima Convertidora de Angiotensina , Corteza Suprarrenal/metabolismo
5.
Neuroradiol J ; : 19714009231224410, 2024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-38171509

RESUMEN

INTRODUCTION: Carotid artery stenting (CAS) through transradial access (TRA) is emerging as an alternative to carotid endarterectomy. However, the current evidence base is limited, mainly comprising single-center studies. OBJECTIVE: This systematic review and meta-analysis aim to assess the safety and effectiveness of TRA for CAS, providing evidence to support clinical decisions. METHODS: We conducted searches on PUBMED, Cochrane Library, Embase, and Web of Science databases, including studies on TRA for CAS. Studies with fewer than 20 patients, non-primary outcomes, and non-full-text articles were excluded. RESULTS: We analyzed 14 studies involving 1,166 patients who underwent CAS via TRA. Procedural success rate was high in 13 studies, with a 95% rate (95% CI; 92%-98%). Crossover to TFA access was observed in 12 studies at 6% (95% CI: 3%-9%). Transradial access failure was reported in four studies, with a rate of 0% (95% CI: 0%-0%). Cannulation failure resulted in a rate of 4% (95% CI: 2%-7%). Asymptomatic radial artery occlusion (ARAO) occurred at a rate of 2% based on eight studies (95% CI: 0%-5%). Forearm hematoma was reported in 10 studies, with an occurrence of 1% (95% CI: 0%-2%). Cerebral vascular attacks (CAV) within 30 days were assessed in 13 studies, indicating a 2% occurrence (95% CI: 1%-2%). CONCLUSION: The findings suggest that TRA for CAS yields promising outcomes with high success rates and low complication rates. Further research should focus on randomized controlled trials and long-term outcomes to validate and extend findings.

6.
Drug Des Devel Ther ; 18: 71-80, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38229917

RESUMEN

Introduction: Nicotine is a major component of cigarette smoke with various detrimental cardiovascular effects, including increased oxidative stress in the heart. Agonism of α2-adrenergic receptors (ARs), such as with dexmedetomidine, has been documented to exert cardioprotective effects against oxidative stress and related apoptosis and necroptosis. α2-ARs are membrane-residing G protein-coupled receptors (GPCRs) that primarily activate Gi/o proteins. They are also subjected to GPCR-kinase (GRK)-2-dependent desensitization, which entails phosphorylation of the agonist-activated receptor by GRK2 to induce its decoupling from G proteins, thus terminating α2AR-mediated G protein signaling. Objective: In the present study, we sought to examine the effects of nicotine on α2AR signaling and effects in H9c2 cardiomyocytes exposed to H2O2 to induce oxidative cellular damage. Methods and Results: As expected, treatment of H9c2 cardiomyocytes with H2O2 significantly decreased cell viability and increased oxidative stress, as assessed by reactive oxygen species (ROS)-associated fluorescence levels (DCF assay) and superoxide dismutase activity. Both H2O2 effects were partly rescued by α2AR activation with brimonidine in control cardiomyocytes but not in cells pretreated with nicotine for 24 hours, in which brimonidine was unable to reduce H2O2-induced cell death and oxidative stress. This was due to severe α2AR desensitization, manifested as very low Gi protein activation by brimonidine, and accompanied by GRK2 upregulation in nicotine-treated cardiomyocytes. Finally, pharmacological inhibition of adenylyl cyclase (AC) blocked H2O2-dependent oxidative damage in nicotine-pretreated H9c2 cardiomyocytes, indicating that α2AR activation protects against oxidative injury via its classic coupling to Gai-mediated AC inhibition. Discussion/Conclusions: Nicotine can negate the cardioprotective effects of α2AR agonists against oxidative injury, which may have important implications for patients treated with this class of drugs that are chronic tobacco smokers.


Asunto(s)
Miocitos Cardíacos , Nicotina , Humanos , Nicotina/farmacología , Nicotina/metabolismo , Peróxido de Hidrógeno/farmacología , Peróxido de Hidrógeno/metabolismo , Receptores Adrenérgicos alfa 2/metabolismo , Estrés Oxidativo , Apoptosis , Tartrato de Brimonidina/metabolismo , Tartrato de Brimonidina/farmacología
7.
Biochem Pharmacol ; 218: 115904, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37922976

RESUMEN

Angiotensin II (AngII), as an octapeptide hormone normally ionized at physiological pH, cannot cross cell membranes and thus, relies on, two (mainly) G protein-coupled receptor (GPCR) types, AT1R and AT2R, to exert its intracellular effects in various organ systems including the cardiovascular one. Although a lot remains to be elucidated about the signaling of the AT2R, AT1R signaling is known to be remarkably versatile, mobilizing a variety of G protein-dependent and independent signal transduction pathways inside cells to produce a biological outcome. Cardiac AT1R signaling leads to hypertrophy, adverse remodeling, fibrosis, while vascular AT1R signaling raises blood pressure via vasoconstriction, but also elicits hypertrophic, vascular growth/proliferation, and pathological remodeling sets of events. In addition, adrenal AT1R is the major physiological stimulus (alongside hyperkalemia) for secretion of aldosterone, a mineralocorticoid hormone that contributes to hypertension, electrolyte abnormalities, and to pathological remodeling of the failing heart. Regulator of G protein Signaling (RGS) proteins, discovered about 25 years ago as GTPase-activating proteins (GAPs) for the Gα subunits of heterotrimeric G proteins, play a central role in silencing G protein signaling from a plethora of GPCRs, including the AngII receptors. Given the importance of AngII and its receptors, but also of several RGS proteins, in cardiovascular homeostasis, the physiological and pathological significance of RGS protein-mediated modulation of cardiovascular AngII signaling comes as no surprise. In the present review, we provide an overview of the current literature on the involvement of RGS proteins in cardiovascular AngII signaling, by discussing their roles in cardiac (cardiomyocyte and cardiofibroblast), vascular (smooth muscle and endothelial cell), and adrenal (medulla and cortex) AngII signaling, separately. Along the way, we also highlight the therapeutic potential of enhancement of, or, in some cases, inhibition of each RGS protein involved in AngII signaling in each one of these cell types.


Asunto(s)
Sistema Cardiovascular , Proteínas RGS , Humanos , Angiotensina II/metabolismo , Sistema Cardiovascular/metabolismo , Proteínas de Unión al GTP/metabolismo , Hipertrofia , Receptor de Angiotensina Tipo 1/metabolismo , Proteínas RGS/genética , Proteínas RGS/metabolismo , Transducción de Señal
8.
J Clin Neurosci ; 117: 104-113, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37788533

RESUMEN

BACKGROUND: Hemangioblastomas (HBs) are highly vascular tumors linked to substantial morbidity and mortality. Recently, interventional neuroradiology has evolved rapidly, spurring interest in preoperative embolization as a possible HB treatment. PURPOSE: This study evaluates the effectiveness and safety of preoperative embolization in managing HB. METHODS: Adhering to PRISMA guidelines, this meta-analysis considered randomized and nonrandomized studies meeting specific criteria, encompassing intracranial HB and preoperative embolization. Primary outcomes were preoperative embolization efficacy and safety. Complications were classified as major (cerebellar ischemia, ischemic strokes, intratumoral hemorrhage, subarachnoid hemorrhage) and minor (transient nystagmus, slight facial nerve palsy, nausea, transient dysarthria, hemiparesis, hemisensory impairment, thrombotic complications, extravasation). RESULTS: Thirteen studies involving 166 patients with preoperative embolization before HB resection were included. Two studies using the Glasgow Outcome Scale (GOS) showed 5 patients with good recovery, 6 with moderate disability, and 3 with severe disability. Major complications occurred in 1% (95% CI: 0% to 3%), and minor complications occurred in 1% (95% CI: 0% to 4%). Intraoperative blood loss during resection was estimated at 464.29 ml (95% CI: 350.63 ml to 614.80 ml). CONCLUSION: Preoperative embolization holds promise in reducing intraoperative bleeding risk in neurosurgical intracranial HB treatment, primarily due to its low complication rates. Nonetheless, additional research and larger-scale studies are essential to establish its long-term efficacy and safety. These findings highlight preoperative embolization as a valuable tool for HB management, potentially enhancing future patient outcomes.


Asunto(s)
Embolización Terapéutica , Hemangioblastoma , Humanos , Hemangioblastoma/terapia , Hemangioblastoma/cirugía , Embolización Terapéutica/efectos adversos , Procedimientos Neuroquirúrgicos , Cuidados Preoperatorios , Pérdida de Sangre Quirúrgica , Resultado del Tratamiento , Estudios Retrospectivos
9.
Eur J Obstet Gynecol Reprod Biol X ; 20: 100234, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37701631

RESUMEN

Objective: The objective of this study is to compare the outcomes of standard cesarean section uterine closure, single-layered running lock sutures, versus using primary horizontal mattress sutures (the K uterine closure technique). Method: This study used a retrospective cohort chart review of patients undergoing elective primary and repeat cesarean section between January 2016 and August 2020 at a South Florida hospital. From 613 included patients, 176 received the K uterine closure technique completed by a triple board-certified physician. Remaining 437 patients received single-layered lock uterine closure completed by other physicians in the same hospital. Clinical, demographic, and post-operative outcomes were collected using patients' operative, post-operative, and progress notes. Results: Using the data gathered, the experimental group demonstrated a greater difference between pre- and post-cesarean section hemoglobin (p < 0.027) and hematocrit (p < 0.014) compared to single-layered lock closure methods. There was a significantly lower estimated blood loss (p < 0.001), however, there was no significant difference in the average quantitative blood loss (p < 0.374). There was also a significant reduction in the length of total and postoperative hospital stay (p < 0.001), but the total operation time was significantly increased (p < 0.016). No significant difference was found in the percentage of patients using opioids as pain management during hospital stay (p < 0.431). There was no need for blood transfusion nor an increase in infectious morbidity using this method. Conclusions: Using the K uterine closure technique for post-cesarean section uterine closure was a reasonable alternative for closure of hysterotomy. It led to decreased duration of hospital stay and no significant difference in quantitative blood loss. The greater difference in hemoglobin and hematocrit for the K uterine closure technique group could be explained due to the significantly greater total operation time. Although the single-layered running lock suture closure is what has been more historically performed by obstetricians for cesarean sections, this data supports the viability and efficacy of the K uterine closure technique as an equally safe, non-inferior alternative. The value of this technique for uterine closure can be confirmed with future prospective studies and potential research in reduction of uterine scar defects. Synopsis: Primary horizontal mattress closure at cesarean section provides a safe alternative to single-layered lock closure and may reduce blood loss, hospital stay, and opioid use.

10.
Interv Neuroradiol ; : 15910199231194665, 2023 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-37593780

RESUMEN

BACKGROUND: The carotid artery stenting (CAS) has two common access sites: transradial access (TRA) and transfemoral access (TFA). However, there's no definitive answer to which one is superior. OBJECTIVE: Compare TRA and TFA for the CAS. METHODS: A systematic review of the literature of studies reporting both TRA and TFA results was conducted following the PRISMA guidelines. PubMed, Cochrane Library, Web of Science and Embase were queried. RESULTS: The meta-analysis examined nine studies comprising 7513 patients who underwent CAS. Of these, 6750 patients had TFA (90%), while 763 had TRA (10%). There was no significant difference in procedure success rates between TRA and TFA, with a risk ratio (RR) of 0.99 (6/9; 95% CI 0.98 to 1.00; I² = 9%, fixed effects). However, cross-over to TFA was more frequent in TRA (odds ratio (OR) 10.37 (6/9; 95% CI 5.18 to 20.77; I² = 17%, fixed effects)). There were no significant differences in terms of major access complications (RR = 0.88 (7/9; 95% CI: 0.29 to 2.63; I² = 0, fixed effects)), total access complications (RR = 1.10 (6/9; 95% CI: 0.56 to 2.15; I² = 7%, fixed effects)), and mean difference in length of stay (Mean difference of -0.08 (3/9; 95% CI -0.18 to 0.02; I² = 0%, fixed effects)). CONCLUSION: There were no significant differences between TFA and TRA in terms of procedure success rates, time, complications, and length of stay, although cross-over to TFA was more common in TRA cases.

11.
Gastroenterol. hepatol. (Ed. impr.) ; 46(5): 360-368, May. 2023. ilus, tab, graf
Artículo en Español | IBECS | ID: ibc-220006

RESUMEN

Antecedentes: El esófago de Barrett (EB) es una entidad con una progresión histológica a malignidad conocida. Los factores de crecimiento insulínico (IGF, de insulin-like growth factor) están involucrados en la carcinogénesis asociada a la obesidad y se han asociado con el riesgo de padecer algunos tipos de cáncer. Objetivos: Evaluar los niveles serológicos de IGF-1 e IGFBP-3 en pacientes con EB y adenocarcinoma de esófago. Pacientes y métodos: Estudio prospectivo de pacientes con EB y adenocarcinoma de esófago explorados con gastroscopia entre septiembre 2012 y diciembre 2015 a los que se realizó una extracción de sangre para la determinación de IGF-1 e IGFBP-3. Se incluyó un grupo control. Resultados: Se incluyeron 116 pacientes: 36 controles, 62 con EB (42 sin displasia y 20 con displasia) y 18 con adenocarcinoma. El IGF-1 y la ratio molar IGF-1/IGFBP-3 presentaron un aumento progresivo en los grupos con EB y adenocarcinoma comparado con los controles (IGF-1: 135,55±66,07ng/ml; 148,33±81,5ng/ml; 108,19±46,69ng/ml, respectivamente; p=0,049) (ratio molar: 0,23±0,91; 0,29±0,11; 0,19±0,06, respectivamente; p=0,001), sin diferencias entre los diferentes grados histológicos. Cincuenta y cuatro de los 65 pacientes con EB fueron seguidos durante una mediana de 58,50 meses (12-113) y 11 de ellos (20,4%) presentaron progresión a displasia de bajo grado (n=8) o displasia de alto grado/adenocarcinoma (n=3), sin encontrar diferencias en el sistema IGF comparado con los que no progresaron. Conclusiones: Los pacientes con EB y adenocarcinoma esofágico presentan cambios en el sistema IGF aunque los niveles de IGF-1 e IGFBP-3 no se correlacionan con la progresión histológica del EB.(AU)


Background: Barrett's esophagus (BE) is an entity with a known histological progression to malignancy. The insulin-like growth factor (IGF) system is involved in the carcinogenesis through obesity-related mechanisms that include IGF and it has been associated with several types of cancer. Objectives: To evaluate the serological levels of IGF-1 and IGFBP-3 in patients with BE and esophageal adenocarcinoma. Patients and methods: Prospective study of patients with BE and esophageal adenocarcinoma who underwent upper endoscopy between September 2012 and December 2015. A baseline determination of IGF-1 and IGFBP-3 was performed. We included a control group of patients without BE. Results: One hundred sixteen patients were included: 36 controls, 62 with BE (42 without dysplasia and 20 with dysplasia) and 18 with adenocarcinoma. IGF-1 and IGF-1/IGFBP-3 molar ratio showed a progression to high levels in BE and adenocarcinoma than in controls (IGF-1: 135.55±66.07ng/ml, 148.33±81.5ng/ml, 108.19±46.69ng/ml, respectively; P=.049) (molar ratio: 0.23±0.91, 0.29±0.11, 0.19±0.06, respectively; P=.001), without differences between the histological types of BE. Fifty-four out of the 65 patients with BE were followed up (median of 58.50 months, range 12–113) and 11 of them (20.4%) presented progression to low-grade dysplasia (n=8) or high-grade dysplasia/adenocarcinoma (n=3), without differences in the IGF system compared with patients without progression. Conclusions: Patients with BE and esophageal adenocarcinoma have changes in the IGF system although the serological levels of IGF-1 and IGFBP-3 do not correlate with histological progression of BE.(AU)


Asunto(s)
Humanos , Esófago de Barrett , Adenocarcinoma , Esófago , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina , Factor I del Crecimiento Similar a la Insulina , Estudios Longitudinales , Estudios Prospectivos
12.
Int J Mol Sci ; 24(7)2023 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-37047106

RESUMEN

The regulator of G protein signaling (RGS) proteins are crucial for the termination of G protein signals elicited by G protein-coupled receptors (GPCRs). This superfamily of cell membrane receptors, by far the largest and most versatile in mammals, including humans, play pivotal roles in the regulation of cardiac function and homeostasis. Perturbations in both the activation and termination of their G protein-mediated signaling underlie numerous heart pathologies, including heart failure (HF) and atrial fibrillation (AFib). Therefore, RGS proteins play important roles in the pathophysiology of these two devasting cardiac diseases, and several of them could be targeted therapeutically. Although close to 40 human RGS proteins have been identified, each RGS protein seems to interact only with a specific set of G protein subunits and GPCR types/subtypes in any given tissue or cell type. Numerous in vitro and in vivo studies in animal models, and also in diseased human heart tissue obtained from transplantations or tissue banks, have provided substantial evidence of the roles various cardiomyocyte RGS proteins play in cardiac normal homeostasis as well as pathophysiology. One RGS protein in particular, RGS4, has been reported in what are now decades-old studies to be selectively upregulated in human HF. It has also been implicated in protection against AFib via knockout mice studies. This review summarizes the current understanding of the functional roles of cardiac RGS proteins and their implications for the treatment of HF and AFib, with a specific focus on RGS4 for the aforementioned reasons but also because it can be targeted successfully with small organic molecule inhibitors.


Asunto(s)
Fibrilación Atrial , Insuficiencia Cardíaca , Proteínas RGS , Animales , Humanos , Ratones , Proteínas de Unión al GTP/metabolismo , Mamíferos/metabolismo , Proteínas RGS/genética , Proteínas RGS/metabolismo , Transducción de Señal/fisiología
13.
Gastroenterol Hepatol ; 46(5): 360-368, 2023 May.
Artículo en Inglés, Español | MEDLINE | ID: mdl-36179948

RESUMEN

BACKGROUND: Barrett's esophagus (BE) is an entity with a known histological progression to malignancy. The insulin-like growth factor (IGF) system is involved in the carcinogenesis through obesity-related mechanisms that include IGF and it has been associated with several types of cancer. OBJECTIVES: To evaluate the serological levels of IGF-1 and IGFBP-3 in patients with BE and esophageal adenocarcinoma. PATIENTS AND METHODS: Prospective study of patients with BE and esophageal adenocarcinoma who underwent upper endoscopy between September 2012 and December 2015. A baseline determination of IGF-1 and IGFBP-3 was performed. We included a control group of patients without BE. RESULTS: One hundred sixteen patients were included: 36 controls, 62 with BE (42 without dysplasia and 20 with dysplasia) and 18 with adenocarcinoma. IGF-1 and IGF-1/IGFBP-3 molar ratio showed a progression to high levels in BE and adenocarcinoma than in controls (IGF-1: 135.55±66.07ng/ml, 148.33±81.5ng/ml, 108.19±46.69ng/ml, respectively; P=.049) (molar ratio: 0.23±0.91, 0.29±0.11, 0.19±0.06, respectively; P=.001), without differences between the histological types of BE. Fifty-four out of the 65 patients with BE were followed up (median of 58.50 months, range 12-113) and 11 of them (20.4%) presented progression to low-grade dysplasia (n=8) or high-grade dysplasia/adenocarcinoma (n=3), without differences in the IGF system compared with patients without progression. CONCLUSIONS: Patients with BE and esophageal adenocarcinoma have changes in the IGF system although the serological levels of IGF-1 and IGFBP-3 do not correlate with histological progression of BE.


Asunto(s)
Adenocarcinoma , Esófago de Barrett , Neoplasias Esofágicas , Humanos , Esófago de Barrett/metabolismo , Esófago de Barrett/patología , Estudios Longitudinales , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina , Factor I del Crecimiento Similar a la Insulina/metabolismo , Estudios Prospectivos , Progresión de la Enfermedad , Neoplasias Esofágicas/patología , Adenocarcinoma/patología
14.
Prog Mol Biol Transl Sci ; 193(1): 145-166, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36357075

RESUMEN

G protein-coupled receptors (GPCRs) play pivotal roles in regulation of cardiovascular homeostasis across all vertebrate species, including humans. In terms of normal cellular function, termination of GPCR signaling via the heterotrimeric G proteins is equally (if not more) important to its stimulation. The Regulator of G protein Signaling (RGS) protein superfamily are indispensable for GPCR signaling cessation at the cell membrane, and thus, for cellular control of GPCR signaling and function. Perturbations in both activation and termination of G protein signaling underlie many examples of cardiovascular dysfunction and heart disease pathogenesis. Despite the plethora of over 30 members comprising the mammalian RGS protein superfamily, each member interacts with a specific set of second messenger pathways and GPCR types/subtypes in a tissue/cell type-specific manner. An increasing number of studies over the past two decades have provided compelling evidence for the involvement of various RGS proteins in physiological regulation of cardiovascular GPCRs and, consequently, also in the pathophysiology of several cardiovascular ailments. This chapter summarizes the current understanding of the functional roles of RGS proteins as they pertain to cardiovascular, i.e., heart, blood vessel, and platelet GPCR function, with a particular focus on their implications for chronic heart failure pathophysiology and therapy.


Asunto(s)
Sistema Cardiovascular , Proteínas de Unión al GTP Heterotriméricas , Proteínas RGS , Humanos , Animales , Proteínas RGS/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Sistema Cardiovascular/metabolismo , Mamíferos/metabolismo
15.
Methods Mol Biol ; 2547: 267-273, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36068469

RESUMEN

α2-Adrenergic receptors (ARs) mediate many cellular actions of epinephrine and norepinephrine, including inhibition of their secretion (sympathetic inhibition) from adrenal chromaffin cells. Like many other G protein-coupled receptors (GPCRs), they undergo agonist-dependent phosphorylation and desensitization by GPCR kinases (GRKs), a phenomenon recently shown to play a major role in the sympathetic overdrive that accompanies and aggravates chronic heart failure. A three-glutamic acid deletion polymorphism in the human α2B-AR subtype gene (Glu301-303) causes impaired agonist-promoted receptor phosphorylation and desensitization, resulting in enhanced signaling to inhibition of cholinergic-induced catecholamine secretion in adrenal chromaffin cells. One of the various pharmacological assays that can be used to quantify and quantitatively compare the degrees of agonist-dependent desensitization, i.e., G protein decoupling, of these two polymorphic α2B-AR variants (or of any two GPCRs for that matter) is the guanosine-5'-O-3-thiotriphosphate (GTPγS) assay that can directly quantify heterotrimeric G protein activation.


Asunto(s)
Proteínas de Unión al GTP Heterotriméricas , Norepinefrina , Epinefrina/farmacología , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Humanos , Norepinefrina/farmacología , Fosforilación
16.
Int J Mol Sci ; 23(10)2022 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-35628613

RESUMEN

Propionic acid is a cell nutrient but also a stimulus for cellular signaling. Free fatty acid receptor (FFAR)-3, also known as GPR41, is a Gi/o protein-coupled receptor (GPCR) that mediates some of the propionate's actions in cells, such as inflammation, fibrosis, and increased firing/norepinephrine release from peripheral sympathetic neurons. The regulator of G-protein Signaling (RGS)-4 inactivates (terminates) both Gi/o- and Gq-protein signaling and, in the heart, protects against atrial fibrillation via calcium signaling attenuation. RGS4 activity is stimulated by ß-adrenergic receptors (ARs) via protein kinase A (PKA)-dependent phosphorylation. Herein, we examined whether RGS4 modulates cardiac FFAR3 signaling/function. We report that RGS4 is essential for dampening of FFAR3 signaling in H9c2 cardiomyocytes, since siRNA-mediated RGS4 depletion significantly enhanced propionate-dependent cAMP lowering, Gi/o activation, p38 MAPK activation, pro-inflammatory interleukin (IL)-1ß and IL-6 production, and pro-fibrotic transforming growth factor (TGF)-ß synthesis. Additionally, catecholamine pretreatment blocked propionic acid/FFAR3 signaling via PKA-dependent activation of RGS4 in H9c2 cardiomyocytes. Finally, RGS4 opposes FFAR3-dependent norepinephrine release from sympathetic-like neurons (differentiated Neuro-2a cells) co-cultured with H9c2 cardiomyocytes, thereby preserving the functional ßAR number of the cardiomyocytes. In conclusion, RGS4 appears essential for propionate/FFAR3 signaling attenuation in both cardiomyocytes and sympathetic neurons, leading to cardioprotection against inflammation/adverse remodeling and to sympatholysis, respectively.


Asunto(s)
Ácidos Grasos no Esterificados , Neuronas , Norepinefrina , Proteínas RGS , Receptores Acoplados a Proteínas G , Señalización del Calcio , Ácidos Grasos no Esterificados/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Humanos , Inflamación/metabolismo , Neuronas/metabolismo , Norepinefrina/metabolismo , Propionatos/metabolismo , Proteínas RGS/genética , Proteínas RGS/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
17.
Int J Mol Sci ; 23(6)2022 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-35328722

RESUMEN

Increasing experimental and clinical evidence points toward a very important role for the gut microbiome and its associated metabolism in human health and disease, including in cardiovascular disorders. Free fatty acids (FFAs) are metabolically produced and utilized as energy substrates during almost every biological process in the human body. Contrary to long- and medium-chain FFAs, which are mainly synthesized from dietary triglycerides, short-chain FFAs (SCFAs) derive from the gut microbiota-mediated fermentation of indigestible dietary fiber. Originally thought to serve only as energy sources, FFAs are now known to act as ligands for a specific group of cell surface receptors called FFA receptors (FFARs), thereby inducing intracellular signaling to exert a variety of cellular and tissue effects. All FFARs are G protein-coupled receptors (GPCRs) that play integral roles in the regulation of metabolism, immunity, inflammation, hormone/neurotransmitter secretion, etc. Four different FFAR types are known to date, with FFAR1 (formerly known as GPR40) and FFAR4 (formerly known as GPR120) mediating long- and medium-chain FFA actions, while FFAR3 (formerly GPR41) and FFAR2 (formerly GPR43) are essentially the SCFA receptors (SCFARs), responding to all SCFAs, including acetic acid, propionic acid, and butyric acid. As with various other organ systems/tissues, the important roles the SCFARs (FFAR2 and FFAR3) play in physiology and in various disorders of the cardiovascular system have been revealed over the last fifteen years. In this review, we discuss the cardiovascular implications of some key (patho)physiological functions of SCFAR signaling pathways, particularly those regulating the neurohormonal control of circulation and adipose tissue homeostasis. Wherever appropriate, we also highlight the potential of these receptors as therapeutic targets for cardiovascular disorders.


Asunto(s)
Receptores de Superficie Celular , Receptores Acoplados a Proteínas G , Ácidos Grasos no Esterificados/metabolismo , Ácidos Grasos Volátiles , Humanos , Receptores de Superficie Celular/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal
18.
J Cardiovasc Pharmacol ; 80(3): 386-392, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34983911

RESUMEN

ABSTRACT: Systolic heart failure (HF) is a chronic clinical syndrome characterized by the reduction in cardiac function and still remains the disease with the highest mortality worldwide. Despite considerable advances in pharmacological treatment, HF represents a severe clinical and social burden. Chronic human HF is characterized by several important neurohormonal perturbations, emanating from both the autonomic nervous system and the adrenal glands. Circulating catecholamines (norepinephrine and epinephrine) and aldosterone elevations are among the salient alterations that confer significant hormonal burden on the already compromised function of the failing heart. This is why sympatholytic treatments (such as ß-blockers) and renin-angiotensin system inhibitors or mineralocorticoid receptor antagonists, which block the effects of angiotensin II (AngII) and aldosterone on the failing heart, are part of the mainstay HF pharmacotherapy presently. The adrenal gland plays an important role in the modulation of cardiac neurohormonal stress because it is the source of almost all aldosterone, of all epinephrine, and of a significant amount of norepinephrine reaching the failing myocardium from the blood circulation. Synthesis and release of these hormones in the adrenals is tightly regulated by adrenal G protein-coupled receptors (GPCRs), such as adrenergic receptors and AngII receptors. In this review, we discuss important aspects of adrenal GPCR signaling and regulation, as they pertain to modulation of cardiac function in the context of chronic HF, by focusing on the 2 best studied adrenal GPCR types in that context, adrenergic receptors and AngII receptors (AT 1 Rs). Particular emphasis is given to findings from the past decade and a half that highlight the emerging roles of the GPCR-kinases and the ß-arrestins in the adrenals, 2 protein families that regulate the signaling and functioning of GPCRs in all tissues, including the myocardium and the adrenal gland.


Asunto(s)
Aldosterona , Insuficiencia Cardíaca , Aldosterona/metabolismo , Epinefrina , Insuficiencia Cardíaca/tratamiento farmacológico , Humanos , Norepinefrina , Receptores Adrenérgicos/uso terapéutico , Receptores Acoplados a Proteínas G/metabolismo
19.
Pharmacol Res ; 174: 105943, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34662735

RESUMEN

The physiological and pathophysiological roles of the angiotensin II type 1 (AT1) receptor, a G protein-coupled receptor ubiquitously expressed throughout the cardiovascular system, have been the focus of intense investigations for decades. The success of angiotensin converting enzyme inhibitors (ACEIs) and of angiotensin receptor blockers (ARBs), which are AT1R-selective antagonists/inverse agonists, in the treatment of heart disease is a testament to the importance of this receptor for cardiovascular homeostasis. Given the pleiotropic signaling of the cardiovascular AT1R and, in an effort to develop yet better drugs for heart disease, the concept of biased signaling has been exploited to design and develop biased AT1R ligands that selectively activate ß-arrestin transduction pathways over Gq protein-dependent pathways. However, by focusing solely on Gq or ß-arrestins, studies on AT1R "biased" signaling & agonism tend to largely ignore other non-Gq-, non ß-arrestin-dependent signaling modalities the very versatile AT1R employs in cardiovascular tissues, including two very important types of signal transducers/regulators: other G protein types (e.g., Gi/o, G12/13) & the Regulator of G protein Signaling (RGS) proteins. In this review, we provide a brief overview of the current state of cardiovascular AT1R biased signaling field with a special focus on the non-Gq-, non ß-arrestin-dependent signaling avenues of this receptor in the cardiovascular system, which usually get left out of the conversation of "biased" AT1R signal transduction.


Asunto(s)
Receptor de Angiotensina Tipo 1/metabolismo , Glándulas Suprarrenales/metabolismo , Animales , Sistema Cardiovascular/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Humanos , Transducción de Señal , beta-Arrestinas/metabolismo
20.
Cells ; 10(6)2021 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-34205363

RESUMEN

The mineralocorticoid aldosterone regulates electrolyte and blood volume homeostasis, but it also adversely modulates the structure and function of the chronically failing heart, through its elevated production in chronic human post-myocardial infarction (MI) heart failure (HF). By activating the mineralocorticoid receptor (MR), a ligand-regulated transcription factor, aldosterone promotes inflammation and fibrosis of the heart, while increasing oxidative stress, ultimately induding mitochondrial dysfunction in the failing myocardium. To reduce morbidity and mortality in advanced stage HF, MR antagonist drugs, such as spironolactone and eplerenone, are used. In addition to the MR, aldosterone can bind and stimulate other receptors, such as the plasma membrane-residing G protein-coupled estrogen receptor (GPER), further complicating it signaling properties in the myocardium. Given the salient role that adrenergic receptor (ARs)-particularly ßARs-play in cardiac physiology and pathology, unsurprisingly, that part of the impact of aldosterone on the failing heart is mediated by its effects on the signaling and function of these receptors. Aldosterone can significantly precipitate the well-documented derangement of cardiac AR signaling and impairment of AR function, critically underlying chronic human HF. One of the main consequences of HF in mammalian models at the cellular level is the presence of mitochondrial dysfunction. As such, preventing mitochondrial dysfunction could be a valid pharmacological target in this condition. This review summarizes the current experimental evidence for this aldosterone/AR crosstalk in both the healthy and failing heart, and the impact of mitochondrial dysfunction in HF. Recent findings from signaling studies focusing on MR and AR crosstalk via non-conventional signaling of molecules that normally terminate the signaling of ARs in the heart, i.e., the G protein-coupled receptor-kinases (GRKs), are also highlighted.


Asunto(s)
Aldosterona/metabolismo , Insuficiencia Cardíaca/metabolismo , Mitocondrias Cardíacas/metabolismo , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Receptores Adrenérgicos/metabolismo , Transducción de Señal , Animales , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/patología , Humanos , Mitocondrias Cardíacas/patología , Infarto del Miocardio/complicaciones , Infarto del Miocardio/patología , Miocardio/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...