Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 5.086
Filtrar
Más filtros

Publication year range
1.
Nature ; 565(7740): 505-510, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30651639

RESUMEN

The increasing prevalence of diabetes has resulted in a global epidemic1. Diabetes is a major cause of blindness, kidney failure, heart attacks, stroke and amputation of lower limbs. These are often caused by changes in blood vessels, such as the expansion of the basement membrane and a loss of vascular cells2-4. Diabetes also impairs the functions of endothelial cells5 and disturbs the communication between endothelial cells and pericytes6. How dysfunction of endothelial cells and/or pericytes leads to diabetic vasculopathy remains largely unknown. Here we report the development of self-organizing three-dimensional human blood vessel organoids from pluripotent stem cells. These human blood vessel organoids contain endothelial cells and pericytes that self-assemble into capillary networks that are enveloped by a basement membrane. Human blood vessel organoids transplanted into mice form a stable, perfused vascular tree, including arteries, arterioles and venules. Exposure of blood vessel organoids to hyperglycaemia and inflammatory cytokines in vitro induces thickening of the vascular basement membrane. Human blood vessels, exposed in vivo to a diabetic milieu in mice, also mimic the microvascular changes found in patients with diabetes. DLL4 and NOTCH3 were identified as key drivers of diabetic vasculopathy in human blood vessels. Therefore, organoids derived from human stem cells faithfully recapitulate the structure and function of human blood vessels and are amenable systems for modelling and identifying the regulators of diabetic vasculopathy, a disease that affects hundreds of millions of patients worldwide.


Asunto(s)
Membrana Basal/patología , Vasos Sanguíneos/patología , Angiopatías Diabéticas/patología , Modelos Biológicos , Organoides/patología , Organoides/trasplante , Proteínas Adaptadoras Transductoras de Señales , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Arterias/citología , Arterias/efectos de los fármacos , Arteriolas/citología , Arteriolas/efectos de los fármacos , Membrana Basal/citología , Membrana Basal/efectos de los fármacos , Vasos Sanguíneos/citología , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/crecimiento & desarrollo , Proteínas de Unión al Calcio , Angiopatías Diabéticas/enzimología , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Humanos , Hiperglucemia/complicaciones , Técnicas In Vitro , Mediadores de Inflamación/farmacología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones , Organoides/citología , Organoides/efectos de los fármacos , Pericitos/citología , Pericitos/efectos de los fármacos , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/efectos de los fármacos , Receptor Notch3/metabolismo , Transducción de Señal , Vénulas/citología , Vénulas/efectos de los fármacos
2.
J Physiol ; 602(14): 3505-3518, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38743485

RESUMEN

NaV1.7 plays a crucial role in inducing and conducting action potentials in pain-transducing sensory nociceptor fibres, suggesting that NaV1.7 blockers could be effective non-opioid analgesics. While SCN9A is expressed in both sensory and autonomic neurons, its functional role in the autonomic system remains less established. Our single neuron rt-PCR analysis revealed that 82% of sympathetic neurons isolated from guinea-pig stellate ganglia expressed NaV1.7 mRNA, with NaV1.3 being the only other tetrodotoxin-sensitive channel expressed in approximately 50% of neurons. We investigated the role of NaV1.7 in conducting action potentials in postganglionic sympathetic nerves and in the sympathetic adrenergic contractions of blood vessels using selective NaV1.7 inhibitors. Two highly selective NaV1.7 blockers, GNE8493 and PF 05089771, significantly inhibited postganglionic compound action potentials by approximately 70% (P < 0.01), with residual activity being blocked by the NaV1.3 inhibitor, ICA 121431. Electrical field stimulation (EFS) induced rapid contractions in guinea-pig isolated aorta, pulmonary arteries, and human isolated pulmonary arteries via stimulation of intrinsic nerves, which were inhibited by prazosin or the NaV1 blocker tetrodotoxin. Our results demonstrated that blocking NaV1.7 with GNE8493, PF 05089771, or ST2262 abolished or strongly inhibited sympathetic adrenergic responses in guinea-pigs and human vascular smooth muscle. These findings support the hypothesis that pharmacologically inhibiting NaV1.7 could potentially reduce sympathetic and parasympathetic function in specific vascular beds and airways. KEY POINTS: 82% of sympathetic neurons isolated from the stellate ganglion predominantly express NaV1.7 mRNA. NaV1.7 blockers inhibit action potential conduction in postganglionic sympathetic nerves. NaV1.7 blockade substantially inhibits sympathetic nerve-mediated adrenergic contractions in human and guinea-pig blood vessels. Pharmacologically blocking NaV1.7 profoundly affects sympathetic and parasympathetic responses in addition to sensory fibres, prompting exploration into the broader physiological consequences of NaV1.7 mutations on autonomic nerve activity.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.7 , Animales , Cobayas , Canal de Sodio Activado por Voltaje NAV1.7/genética , Canal de Sodio Activado por Voltaje NAV1.7/fisiología , Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Humanos , Masculino , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Fibras Simpáticas Posganglionares/fisiología , Fibras Simpáticas Posganglionares/efectos de los fármacos , Femenino , Arterias/fisiología , Arterias/efectos de los fármacos , Arterias/inervación , Bloqueadores de los Canales de Sodio/farmacología , Ganglio Estrellado/fisiología , Sistema Nervioso Simpático/fisiología , Sistema Nervioso Simpático/efectos de los fármacos
3.
Am J Physiol Heart Circ Physiol ; 327(4): H896-H907, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39150393

RESUMEN

Excess sodium consumption contributes to arterial dysfunction in humans. The C57BL/6 strain of mice has been used to identify mechanisms by which arterial dysfunction occurs after excess sodium consumption. However, there are concerns that C57BL/6 mice have strain-specific resistance to high-sodium (HS) diet-induced hypertension. To address this concern, we performed a meta-analysis to determine if excess sodium consumption in C57BL/6 mice induces arterial dysfunction. Databases were searched for HS versus standard diet studies that measured arterial function [i.e., systolic blood pressure (BP), endothelium-dependent dilation (EDD), and central arterial stiffness] in C57BL/6 mice. A total of 39 studies were included, demonstrating that the HS condition resulted in higher systolic BP than control mice with a mean difference of 9.8 mmHg (95% confidence interval [CI] = [5.6, 14], P < 0.001). Subgroup analysis indicated that the systolic BP was higher in HS compared with the control condition when measured during night compared with daytime with telemetry (P < 0.001). We also identified that the difference in systolic BP between HS and control was ∼2.5-fold higher when administered through drinking water than through food (P < 0.001). A total of 12 studies were included, demonstrating that the HS condition resulted in lower EDD than control with a weighted mean difference of -12.0% (95% CI = [-20.0, -4.1], P = 0.003). It should be noted that there was considerable variability across studies with more than half of the studies showing no effect of the HS condition on systolic BP or EDD. In summary, excess sodium consumption elevates systolic BP and impairs EDD in C57BL/6 mice.NEW & NOTEWORTHY C57BL/6 mice are perceived as resistant to high-sodium diet-induced arterial dysfunction. This meta-analysis demonstrates that excess sodium consumption elevates blood pressure and impairs endothelium-dependent dilation in C57BL/6 mice. Nighttime measurements show more pronounced blood pressure elevation. In addition, sodium administration via drinking water, compared with food, induces a greater blood pressure elevation. These findings may be influenced by outlier studies, as the majority of studies showed no adverse effect of excess sodium consumption on arterial function.


Asunto(s)
Ratones Endogámicos C57BL , Sodio en la Dieta , Rigidez Vascular , Animales , Ratones , Arterias/efectos de los fármacos , Arterias/fisiopatología , Arterias/metabolismo , Presión Sanguínea/efectos de los fármacos , Modelos Animales de Enfermedad , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiopatología , Endotelio Vascular/metabolismo , Hipertensión/fisiopatología , Hipertensión/inducido químicamente , Sodio en la Dieta/administración & dosificación , Sodio en la Dieta/efectos adversos , Rigidez Vascular/efectos de los fármacos , Vasodilatación/efectos de los fármacos
4.
Pediatr Res ; 95(7): 1758-1763, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38310195

RESUMEN

BACKGROUND: Perinatal hypoxia affects a lot of neonates worldwide every year, however its effects on the functioning of systemic circulation are not clear yet. We aimed at investigation the effects of perinatal hypoxia on the second day of life on the functioning of the rat systemic vasculature in early postnatal period. METHODS: 2-day-old male rat pups were exposed to normobaric hypoxia (8% O2, 92% N2) for 2 hours. At the 11-14 days cutaneous (saphenous) arteries were isolated and studied by wire myography and Western blotting. RESULTS: Hypoxia weakened the contribution of anticontractile influence of NO, but did not affect the contribution of Rho-kinase or Kv7 channels to the contraction to α1-adrenergic agonist methoxamine. The content of eNOS and protein kinase G were not altered by hypoxic conditions. CONCLUSION: Perinatal hypoxia in rats at the second day of life leads to the decrease of anticontractile effect of NO in the systemic arteries in early postnatal ontogenesis (at the age of 11-14 days). Decreased anticontractile effect of NO can be the reason for insufficient blood supply and represent a risk factor for the development of cardiovascular disorders. IMPACT: The mechanisms of perinatal hypoxia influences on systemic circulation are almost unknown. We have shown that perinatal hypoxia weakens anticontractile influence of nitric oxide in early postnatal period. The influence of perinatal hypoxia on systemic circulation should be taken into account during treatment of newborns suffered from the lack of oxygen.


Asunto(s)
Animales Recién Nacidos , Arterias , Hipoxia , Óxido Nítrico , Animales , Óxido Nítrico/metabolismo , Masculino , Ratas , Arterias/efectos de los fármacos , Arterias/crecimiento & desarrollo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Vasoconstricción/efectos de los fármacos , Ratas Wistar , Metoxamina/farmacología , Quinasas Asociadas a rho/metabolismo
5.
Ecotoxicol Environ Saf ; 285: 117083, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-39317073

RESUMEN

Humans are ubiquitously exposed to crotonaldehyde (CRA) endogenously and exogenously. Deeper knowledge of the pharmacological and toxicological characteristics and the mechanisms of CRA on vasculature is urgently needed for prevention of its harmfulness. The effects of acute and prolonged exposure to CRA were studied in rat isolated arteries and arterial smooth muscle cells (ASMCs). Instant exposure to CRA (1-300 µM) concentration-dependently declined the tension of pre-constricted arteries with an irreversible depression on the contractility. Prolonged exposure of rat coronary arteries (RCAs) to CRA concentration- and time-dependently depressed the arterial contractile responsiveness to various vasoconstrictors including depolarization, U46619, serotonin and Bay K8644 (an agonist of voltage-gated Ca2+ channels (VGCCs)). In fresh RCA ASMCs, CRA abated depolarization-induced elevation of intracellular Ca2+ ([Ca2+]i). Electrophysiological study revealed that acute exposure to CRA depressed the functions of Ca2+-activated Cl- channels (CaCCs), voltage-gated K+ (Kv) channels and inward rectifier K+ (Kir) channels in RCA ASMCs. Prolonged exposure of RCAs to CRA reduced the expressions of these ion channels in RCA ASMCs, disordered tissue frames, injured arterial cells, and increased autophagosomes in both ASMCs and endothelial cells. In rat aortic smooth muscle cells (A7r5), CRA exposure decreased the cell viability, elevated the intracellular levels of reactive oxygen species, reduced the mitochondrial membrane potential, and enhanced autophagy. Taken together, the present study for the first time portrays a clearer panoramic outline of the vascular effects and the mechanisms of CRA on arteries, demonstrates that CRA impairs arterial contractility, depresses VGCCs, CaCCs, Kv channels and Kir channels, reduces cell viability, and destroys the arterial histiocytes, and suggests that excessive oxidative stress, mitochondrial dysfunction and autophagy underlie these vascular damages. These findings are significant for the comprehensive evaluation of the vicious effects of CRA on arteries and suggest potential preventive strategies.


Asunto(s)
Aldehídos , Autofagia , Mitocondrias , Miocitos del Músculo Liso , Especies Reactivas de Oxígeno , Animales , Especies Reactivas de Oxígeno/metabolismo , Autofagia/efectos de los fármacos , Ratas , Mitocondrias/efectos de los fármacos , Masculino , Miocitos del Músculo Liso/efectos de los fármacos , Aldehídos/toxicidad , Músculo Liso Vascular/efectos de los fármacos , Ratas Sprague-Dawley , Calcio/metabolismo , Arterias/efectos de los fármacos
6.
Int J Mol Sci ; 25(13)2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-39000253

RESUMEN

It has been reported that, in the spontaneously hypertensive rat (SHR) model of hypertension, different components of the G-protein/adenylate cyclase (AC)/Calcium-activated potassium channel of high conductance (BK) channel signaling pathway are altered differently. In the upstream part of the pathway (G-protein/AC), a comparatively low efficacy has been established, whereas downstream BK currents seem to be increased. Thus, the overall performance of this signaling pathway in SHR is elusive. For a better understanding, we focused on one aspect, the direct targeting of the BK channel by the G-protein/AC pathway and tested the hypothesis that the comparatively low AC pathway efficacy in SHR results in a reduced agonist-induced stimulation of BK currents. This hypothesis was investigated using freshly isolated smooth muscle cells from WKY and SHR rat tail artery and the patch-clamp technique. It was observed that: (1) single BK channels have similar current-voltage relationships, voltage-dependence and calcium sensitivity; (2) BK currents in cells with a strong buffering of the BK channel activator calcium have similar current-voltage relationships; (3) the iloprost-induced concentration-dependent increase of the BK current is larger in WKY compared to SHR; (4) the effects of activators of the PKA pathway, the catalytic subunit of PKA and the potent and selective cAMP-analogue Sp-5,6-DCl-cBIMPS on BK currents are similar. Thus, our data suggest that the lower iloprost-induced stimulation of the BK current in freshly isolated rat tail artery smooth muscle cells from SHR compared with WKY is due to the lower efficacy of upstream elements of the G-Protein/AC/BK channel pathway.


Asunto(s)
Calcio , Hipertensión , Iloprost , Canales de Potasio de Gran Conductancia Activados por el Calcio , Músculo Liso Vascular , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Vasodilatadores , Animales , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/citología , Ratas , Calcio/metabolismo , Iloprost/farmacología , Hipertensión/metabolismo , Hipertensión/tratamiento farmacológico , Vasodilatadores/farmacología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Masculino , Arterias/efectos de los fármacos , Arterias/metabolismo , Cola (estructura animal)/irrigación sanguínea , Transducción de Señal/efectos de los fármacos
7.
Am J Physiol Endocrinol Metab ; 322(2): E173-E180, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34957859

RESUMEN

Microvascular insulin resistance is present in metabolic syndrome and may contribute to increased cardiovascular disease risk and the impaired metabolic response to insulin observed. Metformin improves metabolic insulin resistance in humans. Its effects on macro and microvascular insulin resistance have not been defined. Eleven subjects with nondiabetic metabolic syndrome were studied four times (before and after 12 wk of treatment with placebo or metformin) using a crossover design, with an 8-wk washout interval between treatments. On each occasion, we measured three indices of large artery function [pulse wave velocity (PWV), radial pulse wave separation analysis (PWSA), brachial artery endothelial function (flow-mediated dilation-FMD)] as well as muscle microvascular perfusion [contrast-enhanced ultrasound (CEU)] before and at 120 min into a 150 min, 1 mU/min/kg euglycemic insulin clamp. Metformin decreased body mass index (BMI), fat weight, and % body fat (P < 0.05, each), however, placebo had no effect. Metformin (not placebo) improved metabolic insulin sensitivity, (clamp glucose infusion rate, P < 0.01), PWV, and FMD after insulin were unaffected by metformin treatment. PWSA improved with insulin only after metformin P < 0.01). Insulin decreased muscle microvascular blood volume measured by contrast ultrasound both before and after placebo and before metformin (P < 0.02 for each) but not after metformin. Short-term metformin treatment improves both metabolic and muscle microvascular response to insulin. Metformin's effect on microvascular insulin responsiveness may contribute to its beneficial metabolic effects. Metformin did not improve aortic stiffness or brachial artery endothelial function, but enhanced radial pulse wave properties consistent with relaxation of smaller arterioles.NEW & NOTEWORTHY Metformin, a first-line treatment for type 2 diabetes, is often used in patients with insulin resistance and metabolic syndrome. Here, we provide the first evidence for metformin improving muscle microvascular insulin sensitivity in insulin-resistant humans. Simultaneously, metformin improved muscle glucose disposal, supporting a close relationship between insulin's microvascular and its metabolic actions in muscle. Whether enhanced microvascular insulin sensitivity contributes to metformin's ability to decrease microvascular complications in diabetes remains to be resolved.


Asunto(s)
Hipoglucemiantes/administración & dosificación , Resistencia a la Insulina , Síndrome Metabólico/tratamiento farmacológico , Síndrome Metabólico/metabolismo , Metformina/administración & dosificación , Microcirculación/efectos de los fármacos , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/metabolismo , Arterias/efectos de los fármacos , Arterias/metabolismo , Velocidad del Flujo Sanguíneo/efectos de los fármacos , Glucemia/metabolismo , Índice de Masa Corporal , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Femenino , Técnica de Clampeo de la Glucosa , Humanos , Insulina/administración & dosificación , Insulina/metabolismo , Masculino , Persona de Mediana Edad , Análisis de la Onda del Pulso , Distribución Aleatoria , Resultado del Tratamiento , Rigidez Vascular/efectos de los fármacos
8.
Prostate ; 82(1): 13-25, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34570375

RESUMEN

INTRODUCTION: Androgen deprivation therapy (ADT) is a key treatment modality in the management of prostate cancer (PCa), especially for patients with metastatic disease. Increasing evidences suggest that patients who received ADT have increased incidence of diabetes, myocardial infarction, stroke, and even mortality. It is important to understand the pathophysiological mechanisms on how ADT increases cardiovascular risk and induces cardiovascular events, which would provide important information for potential implementation of preventive measures. METHODS: Twenty-six 12-week-old male SD rats were divided into four groups for different types of ADTs including: the bilateral orchidectomy group (Orx), LHRH agonist group (leuprolide), LHRH antagonist group (degarelix), and control group. After treated with drug or adjuvant injection every 3 weeks for 24 weeks, all rats were sacrificed and total blood were collected. Aorta, renal arteries, and kidney were preserved for functional assay, immunohistochemistry, western blot, and quantitative reverse-transcription polymerase chain reaction. RESULTS: In vascular reactivity assays, aorta, intrarenal, and coronary arteries of all three ADT groups showed endothelial dysfunction. AT1R and related molecules at protein and messenger RNA (mRNA) level were tested, and AT1R pathway was shown to be activated and played a role in endothelial dysfunction. Both ACE and AT1R mRNA levels were doubled in the aorta in the leuprolide group while Orx and degarelix groups showed upregulation of AT1R in the kidney tissues. By immunohistochemistry, our result showed higher expression of AT1R in the intrarenal arteries of leuprolide and degarelix groups. The role of reactive oxygen species in endothelial dysfunction was confirmed by DHE fluorescence, nitrotyrosine overexpression, and upregulation of NOX2 in the different ADT treatment groups. CONCLUSION: ADT causes endothelial dysfunction in male rats. GnRH receptor agonist compared to GnRH receptor antagonist, showed more impairment of endothelial function in the aorta and intrarenal arteries. Such change might be associated with upregulation and activation of AngII-AT1R-NOX2 induced oxidative stress in the vasculature. These results help to explain the different cardiovascular risks and outcomes related to different modalities of ADT treatment.


Asunto(s)
Antagonistas de Andrógenos , Arterias , Endotelio Vascular , Leuprolida , Oligopéptidos , Orquiectomía/métodos , Antagonistas de Andrógenos/efectos adversos , Antagonistas de Andrógenos/análisis , Antagonistas de Andrógenos/metabolismo , Animales , Arterias/efectos de los fármacos , Arterias/metabolismo , Arterias/patología , Correlación de Datos , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Hormona Liberadora de Gonadotropina/agonistas , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Factores de Riesgo de Enfermedad Cardiaca , Inmunohistoquímica , Leuprolida/administración & dosificación , Leuprolida/efectos adversos , Oligopéptidos/administración & dosificación , Oligopéptidos/efectos adversos , Ratas , Especies Reactivas de Oxígeno/análisis , Receptor de Angiotensina Tipo 1/análisis , Receptor de Angiotensina Tipo 1/metabolismo
9.
Circ Res ; 126(9): 1178-1189, 2020 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-32324506

RESUMEN

The potential of CD31 as a therapeutic target in atherosclerosis has been considered ever since its cloning in the 1990s, but the exact role played by this molecule in the biologic events underlying atherosclerosis has remained controversial, resulting in the stalling of any therapeutic perspective. Due to the supposed cell adhesive properties of CD31, specific monoclonal antibodies and recombinant proteins were regarded as blocking agents because their use prevented the arrival of leukocytes at sites of acute inflammation. However, the observed effect of those compounds likely resulted from the engagement of the immunomodulatory function of CD31 signaling. This was acknowledged only later though, upon the discovery of CD31's 2 intracytoplasmic tyrosine residues called immunoreceptor tyrosine inhibitory motifs. A growing body of evidence currently points at a therapeutic potential for CD31 agonists in atherothrombosis. Clinical observations show that CD31 expression is altered at the surface of leukocytes infiltrating unhealed atherothrombotic lesions and that the physiological immunomodulatory functions of CD31 are lost at the surface of blood leukocytes in patients with acute coronary syndromes. On the contrary, translational studies using candidate therapeutic molecules in laboratory animals have provided encouraging results: synthetic peptides administered to atherosclerotic mice as systemic drugs in the acute phases of atherosclerotic complications favor the healing of wounded arteries, whereas the immobilization of CD31 agonist peptides onto coronary stents implanted in farm pigs favors their peaceful integration within the coronary arterial wall.


Asunto(s)
Arterias/efectos de los fármacos , Aterosclerosis/terapia , Fármacos Cardiovasculares/uso terapéutico , Enfermedad de la Arteria Coronaria/terapia , Stents Liberadores de Fármacos , Factores Inmunológicos/uso terapéutico , Intervención Coronaria Percutánea/instrumentación , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/agonistas , Animales , Arterias/inmunología , Arterias/metabolismo , Arterias/patología , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Enfermedad de la Arteria Coronaria/inmunología , Enfermedad de la Arteria Coronaria/metabolismo , Enfermedad de la Arteria Coronaria/patología , Humanos , Terapia Molecular Dirigida , Placa Aterosclerótica , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Diseño de Prótesis , Transducción de Señal
10.
Circ Res ; 126(12): 1746-1759, 2020 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-32279581

RESUMEN

RATIONALE: Dysregulated purinergic signaling transduction plays important roles in the pathogenesis of cardiovascular diseases. However, the role and mechanism of vascular smooth muscle cell (VSMC)-released ATP in the regulation of blood pressure, and the pathogenesis of hypertension remain unknown. FAM3A (family with sequence similarity 3 member A) is a new mitochondrial protein that enhances ATP production and release. High expression of FAM3A in VSMC suggests it may play a role in regulating vascular constriction and blood pressure. OBJECTIVE: To determine the role and mechanism of FAM3A-ATP signaling pathway in VSMCs in the regulation of blood pressure and the pathogenesis of hypertension. METHODS AND RESULTS: In the media layer of hypertensive rat and mouse arteries, and the internal mammary artery of hypertensive patients, FAM3A expression was increased. VSMC-specific deletion of FAM3A reduced vessel contractility and blood pressure levels in mice. Moreover, deletion of FAM3A in VSMC attenuated Ang II (angiotensin II)-induced vascular constriction and remodeling, hypertension, and cardiac hypertrophy in mice. In cultured VSMCs, Ang II activated HSF1 (heat shock factor 1) to stimulate FAM3A expression, activating ATP-P2 receptor pathway to promote the change of VSMCs from contractile phenotype to proliferative phenotype. In the VSMC layer of spontaneously hypertensive rat arteries, Ang II-induced hypertensive mouse arteries and the internal mammary artery of hypertensive patients, HSF1 expression was increased. Treatment with HSF1 inhibitor reduced artery contractility and ameliorated hypertension of spontaneously hypertensive rats. CONCLUSIONS: FAM3A is an important regulator of vascular constriction and blood pressure. Overactivation of HSF1-FAM3A-ATP signaling cascade in VSMCs plays important roles in Ang II-induced hypertension and cardiovascular diseases. Inhibitors of HSF1 could be potentially used to treat hypertension.


Asunto(s)
Cardiomegalia/metabolismo , Citocinas/metabolismo , Hipertensión/metabolismo , Músculo Liso Vascular/metabolismo , Adenosina Trifosfato/metabolismo , Angiotensina II/farmacología , Animales , Arterias/efectos de los fármacos , Arterias/metabolismo , Arterias/fisiopatología , Presión Sanguínea , Cardiomegalia/fisiopatología , Células Cultivadas , Citocinas/genética , Femenino , Eliminación de Gen , Factores de Transcripción del Choque Térmico/metabolismo , Humanos , Hipertensión/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/fisiopatología , Miocitos del Músculo Liso/metabolismo , Ratas , Receptores Purinérgicos P2/metabolismo , Vasoconstricción , Vasoconstrictores/farmacología
11.
Circ Res ; 127(6): 811-823, 2020 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-32546048

RESUMEN

RATIONALE: Arterial inflammation manifested as atherosclerosis is the leading cause of mortality worldwide. Genome-wide association studies have identified a prominent role of HDAC (histone deacetylase)-9 in atherosclerosis and its clinical complications including stroke and myocardial infarction. OBJECTIVE: To determine the mechanisms linking HDAC9 to these vascular pathologies and explore its therapeutic potential for atheroprotection. METHODS AND RESULTS: We studied the effects of Hdac9 on features of plaque vulnerability using bone marrow reconstitution experiments and pharmacological targeting with a small molecule inhibitor in hyperlipidemic mice. We further used 2-photon and intravital microscopy to study endothelial activation and leukocyte-endothelial interactions. We show that hematopoietic Hdac9 deficiency reduces lesional macrophage content while increasing fibrous cap thickness thus conferring plaque stability. We demonstrate that HDAC9 binds to IKK (inhibitory kappa B kinase)-α and ß, resulting in their deacetylation and subsequent activation, which drives inflammatory responses in both macrophages and endothelial cells. Pharmacological inhibition of HDAC9 with the class IIa HDAC inhibitor TMP195 attenuates lesion formation by reducing endothelial activation and leukocyte recruitment along with limiting proinflammatory responses in macrophages. Transcriptional profiling using RNA sequencing revealed that TMP195 downregulates key inflammatory pathways consistent with inhibitory effects on IKKß. TMP195 mitigates the progression of established lesions and inhibits the infiltration of inflammatory cells. Moreover, TMP195 diminishes features of plaque vulnerability and thereby enhances plaque stability in advanced lesions. Ex vivo treatment of monocytes from patients with established atherosclerosis reduced the production of inflammatory cytokines including IL (interleukin)-1ß and IL-6. CONCLUSIONS: Our findings identify HDAC9 as a regulator of atherosclerotic plaque stability and IKK activation thus providing a mechanistic explanation for the prominence of HDAC9 as a vascular risk locus in genome-wide association studies. Its therapeutic inhibition may provide a potent lever to alleviate vascular inflammation. Graphical Abstract: A graphical abstract is available for this article.


Asunto(s)
Arterias/enzimología , Aterosclerosis/enzimología , Histona Desacetilasas/metabolismo , Quinasa I-kappa B/metabolismo , Placa Aterosclerótica , Proteínas Represoras/metabolismo , Acetilación , Anciano , Anciano de 80 o más Años , Animales , Arterias/efectos de los fármacos , Arterias/patología , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/genética , Aterosclerosis/patología , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/enzimología , Células Endoteliales/patología , Activación Enzimática , Femenino , Fibrosis , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/genética , Humanos , Quinasa I-kappa B/genética , Mediadores de Inflamación/metabolismo , Rodamiento de Leucocito , Macrófagos/enzimología , Macrófagos/patología , Masculino , Ratones Noqueados para ApoE , Persona de Mediana Edad , Monocitos/enzimología , Monocitos/patología , Unión Proteica , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Transducción de Señal
13.
Arterioscler Thromb Vasc Biol ; 41(1): 11-19, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33232199

RESUMEN

Mineralization of cardiovascular structures including blood vessels and heart valves is a common feature. We postulate that ectopic mineralization is a response-to-injury in which signals delivered to cells trigger a chain of events to restore and repair tissues. Maladaptive response to external or internal signals promote the expression of danger-associated molecular patterns, which, in turn, promote, when expressed chronically, a procalcifying gene program. Growing evidence suggest that danger-associated molecular patterns such as oxyphospholipids and small lipid mediators, generated by enzyme activity, are involved in the transition of vascular smooth muscle cells and valve interstitial cells to an osteoblast-like phenotype. Understanding the regulation and the molecular processes underpinning the mineralization of atherosclerotic plaques and cardiac valves are providing valuable mechanistic insights, which could lead to the development of novel therapies. Herein, we provide a focus account on the role oxyphospholipids and their mediators in the development of mineralization in plaques and calcific aortic valve disease.


Asunto(s)
Estenosis de la Válvula Aórtica/metabolismo , Válvula Aórtica/metabolismo , Válvula Aórtica/patología , Arterias/metabolismo , Calcinosis/metabolismo , Fosfolípidos/metabolismo , Calcificación Vascular/metabolismo , Animales , Válvula Aórtica/efectos de los fármacos , Estenosis de la Válvula Aórtica/tratamiento farmacológico , Estenosis de la Válvula Aórtica/patología , Arterias/efectos de los fármacos , Arterias/patología , Calcinosis/tratamiento farmacológico , Calcinosis/patología , Plasticidad de la Célula , Humanos , Oxidación-Reducción , Placa Aterosclerótica , Transducción de Señal , Calcificación Vascular/tratamiento farmacológico , Calcificación Vascular/patología
14.
Arterioscler Thromb Vasc Biol ; 41(1): 117-127, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33115271

RESUMEN

Cardiovascular calcification is an insidious form of ectopic tissue mineralization that presents as a frequent comorbidity of atherosclerosis, aortic valve stenosis, diabetes, renal failure, and chronic inflammation. Calcification of the vasculature and heart valves contributes to mortality in these diseases. An inability to clinically image or detect early microcalcification coupled with an utter lack of pharmaceutical therapies capable of inhibiting or regressing entrenched and detectable macrocalcification has led to a prominent and deadly gap in care for a growing portion of our rapidly aging population. Recognition of this mounting concern has arisen over the past decade and led to a series of revolutionary works that has begun to pull back the curtain on the pathogenesis, mechanistic basis, and causative drivers of cardiovascular calcification. Central to this progress is the discovery that calcifying extracellular vesicles act as active precursors of cardiovascular microcalcification in diverse vascular beds. More recently, the omics revolution has resulted in the collection and quantification of vast amounts of molecular-level data. As the field has become poised to leverage these resources for drug discovery, new means of deriving relevant biological insights from these rich and complex datasets have come into focus through the careful application of systems biology and network medicine approaches. As we look onward toward the next decade, we envision a growing need to standardize approaches to study this complex and multifaceted clinical problem and expect that a push to translate mechanistic findings into therapeutics will begin to finally provide relief for those impacted by this disease.


Asunto(s)
Estenosis de la Válvula Aórtica/metabolismo , Válvula Aórtica/metabolismo , Válvula Aórtica/patología , Arterias/metabolismo , Calcinosis/metabolismo , Vesículas Extracelulares/metabolismo , Osteogénesis , Calcificación Vascular/metabolismo , Animales , Válvula Aórtica/efectos de los fármacos , Estenosis de la Válvula Aórtica/tratamiento farmacológico , Estenosis de la Válvula Aórtica/genética , Estenosis de la Válvula Aórtica/patología , Arterias/efectos de los fármacos , Arterias/patología , Calcinosis/tratamiento farmacológico , Calcinosis/genética , Calcinosis/patología , Desarrollo de Medicamentos , Descubrimiento de Drogas , Vesículas Extracelulares/patología , Regulación de la Expresión Génica , Genómica , Humanos , Osteogénesis/efectos de los fármacos , Transducción de Señal , Calcificación Vascular/tratamiento farmacológico , Calcificación Vascular/genética , Calcificación Vascular/patología
15.
Int J Mol Sci ; 23(4)2022 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-35216422

RESUMEN

Arterial calcification is a common feature of pseudoxanthoma elasticum (PXE), a disease characterized by ABCC6 mutations, inducing a deficiency in pyrophosphate, a key inhibitor of calcium phosphate crystallization in arteries. METHODS: we analyzed whether long-term exposure of Abcc6-/- mice (a murine model of PXE) to a mild vitamin D supplementation, with or without calcium, would impact the development of vascular calcification. Eight groups of mice (including Abcc6-/- and wild-type) received vitamin D supplementation every 2 weeks, a calcium-enriched diet alone (calcium in drinking water), both vitamin D supplementation and calcium-enriched diet, or a standard diet (controls) for 6 months. Aorta and kidney artery calcification was assessed by 3D-micro-computed tomography, Optical PhotoThermal IR (OPTIR) spectroscopy, scanning electron microscopy coupled with energy-dispersive X-ray spectroscopy (SEM-EDS) and Yasue staining. RESULTS: at 6 months, although vitamin D and/or calcium did not significantly increase serum calcium levels, vitamin D and calcium supplementation significantly worsened aorta and renal artery calcification in Abcc6-/- mice. CONCLUSIONS: vitamin D and/or calcium supplementation accelerate vascular calcification in a murine model of PXE. These results sound a warning regarding the use of these supplementations in PXE patients and, to a larger extent, patients with low systemic pyrophosphate levels.


Asunto(s)
Calcificación Fisiológica/efectos de los fármacos , Calcio de la Dieta/farmacología , Calcio/farmacología , Seudoxantoma Elástico/tratamiento farmacológico , Calcificación Vascular/tratamiento farmacológico , Vitamina D/farmacología , Animales , Arterias/efectos de los fármacos , Arterias/metabolismo , Suplementos Dietéticos , Modelos Animales de Enfermedad , Femenino , Ratones , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Seudoxantoma Elástico/metabolismo , Calcificación Vascular/metabolismo
16.
Int J Mol Sci ; 23(3)2022 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-35163382

RESUMEN

Transient receptor potential melastatin-4 (TRPM4) is activated by an increase in intracellular Ca2+ concentration and is expressed on smooth muscle cells (SMCs). It is implicated in the myogenic constriction of cerebral arteries. We hypothesized that TRPM4 has a general role in intracellular Ca2+ signal amplification in a wide range of blood vessels. TRPM4 function was tested with the TRPM4 antagonist 9-phenanthrol and the TRPM4 activator A23187 on the cardiovascular responses of the rat, in vivo and in isolated basilar, mesenteric, and skeletal muscle arteries. TRPM4 inhibition by 9-phenanthrol resulted in hypotension and a decreased heart rate in the rat. TRPM4 inhibition completely antagonized myogenic tone development and norepinephrine-evoked vasoconstriction, and depolarization (high extracellular KCl concentration) evoked vasoconstriction in a wide range of peripheral arteries. Vasorelaxation caused by TRPM4 inhibition was accompanied by a significant decrease in intracellular Ca2+ concentration, suggesting an inhibition of Ca2+ signal amplification. Immunohistochemistry confirmed TRPM4 expression in the smooth muscle cells of the peripheral arteries. Finally, TRPM4 activation by the Ca2+ ionophore A23187 was competitively inhibited by 9-phenanthrol. In summary, TRPM4 was identified as an essential Ca2+-amplifying channel in peripheral arteries, contributing to both myogenic tone and agonist responses. These results suggest an important role for TRPM4 in the circulation. The modulation of TRPM4 activity may be a therapeutic target for hypertension. Furthermore, the Ca2+ ionophore A23187 was identified as the first high-affinity (nanomolar) direct activator of TRPM4, acting on the 9-phenanthrol binding site.


Asunto(s)
Señalización del Calcio , Canales Catiónicos TRPM/metabolismo , Vasoconstricción , Administración Intravenosa , Animales , Arterias/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Calcimicina/farmacología , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Frecuencia Cardíaca/efectos de los fármacos , Ionóforos/farmacología , Masculino , Desarrollo de Músculos/efectos de los fármacos , Músculo Esquelético/irrigación sanguínea , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Norepinefrina/farmacología , Fenantrenos/administración & dosificación , Fenantrenos/farmacología , Cloruro de Potasio/farmacología , Ratas Wistar , Canales Catiónicos TRPM/agonistas , Vasoconstricción/efectos de los fármacos
17.
Mol Pharmacol ; 100(3): 271-282, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34330822

RESUMEN

G protein-coupled receptor 30 (GPR30) is a membrane receptor reported to bind 17ß-estradiol (E2) and mediate rapid nongenomic estrogen responses, hence also named G protein-coupled estrogen receptor. G-1 is a proposed GPR30-specific agonist that has been used to implicate the receptor in several pathophysiological events. However, controversy surrounds the role of GPR30 in G-1 and E2 responses. We investigated GPR30 activity in the absence and presence of G-1 and E2 in several eukaryotic systems ex vivo and in vitro in the absence and presence of the receptor. Ex vivo activity was addressed using the caudal artery from wild-type (WT) and GPR30 knockout (KO) mice, and in vitro activity was addressed using a HeLa cell line stably expressing a synthetic multifunctional promoter (nuclear factor κB, signal transducer and activator of transcription, activator protein 1)-luciferase construct (HFF11 cells) and a human GPR30-inducible T-REx system (T-REx HFF11 cells), HFF11 and human embryonic kidney 293 cells transiently expressing WT GPR30 and GPR30 lacking the C-terminal PDZ (postsynaptic density-95/discs-large /zonula occludens-1 homology) motif SSAV, and yeast Saccharomyces cerevisiae transformed to express GPR30. WT and KO arteries exhibited similar contractile responses to 60 mM KCl and 0.3 µM cirazoline, and G-1 relaxed both arteries with the same potency and efficacy. Furthermore, expression of GPR30 did not introduce any responses to 1 µM G-1 and 0.1 µM E2 in vitro. On the other hand, receptor expression caused considerable ligand-independent activity in vitro, which was receptor PDZ motif-dependent in mammalian cells. We conclude from these results that GPR30 exhibits ligand-independent activity in vitro but no G-1- or E2-stimulated activity in any of the systems used. SIGNIFICANCE STATEMENT: Much controversy surrounds 17ß-estradiol (E2) and G-1 as G protein-coupled receptor 30 (GPR30) agonists. We used several recombinant eukaryotic systems ex vivo and in vitro with and without GPR30 expression to address the role of this receptor in responses to these proposed agonists. Our results show that GPR30 exhibits considerable ligand-independent activity in vitro but no G-1- or E2-stimulated activity in any of the systems used. Thus, classifying GPR30 as an estrogen receptor and G-1 as a specific GPR30 agonist is unfounded.


Asunto(s)
Ciclopentanos/farmacología , Estradiol/farmacología , Quinolinas/farmacología , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Arterias/efectos de los fármacos , Línea Celular , Homólogo 4 de la Proteína Discs Large/metabolismo , Femenino , Humanos , Ligandos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Relajación Muscular/efectos de los fármacos , Dominios PDZ/genética , Receptores de Estrógenos/efectos de los fármacos , Receptores de Estrógenos/genética , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores Acoplados a Proteínas G/genética , Saccharomyces cerevisiae/genética
18.
J Cell Physiol ; 236(4): 2333-2351, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32875580

RESUMEN

Proprotein convertase subtilisin/kexin type-9 (PCSK9), a member of the proprotein convertase family, is an important drug target because of its crucial role in lipid metabolism. Emerging evidence suggests a direct role of localized PCSK9 in the pathogenesis of vascular diseases. With this in our consideration, we reviewed PCSK9 physiology with respect to recent development and major studies (clinical and experimental) on PCSK9 functionality in vascular disease. PCSK9 upregulates low-density lipoprotein (LDL)-cholesterol levels by binding to the LDL-receptor (LDLR) and facilitating its lysosomal degradation. PCSK9 gain-of-function mutations have been confirmed as a novel genetic mechanism for familial hypercholesterolemia. Elevated serum PCSK9 levels in patients with vascular diseases may contribute to coronary artery disease, atherosclerosis, cerebrovascular diseases, vasculitis, aortic diseases, and arterial aging pathogenesis. Experimental models of atherosclerosis, arterial aneurysm, and coronary or carotid artery ligation also support PCSK9 contribution to inflammatory response and disease progression, through LDLR-dependent or -independent mechanisms. More recently, several clinical trials have confirmed that anti-PCSK9 monoclonal antibodies can reduce systemic LDL levels, total nonfatal cardiovascular events, and all-cause mortality. Interaction of PCSK9 with other receptor proteins (LDLR-related proteins, cluster of differentiation family members, epithelial Na+ channels, and sortilin) may underlie its roles in vascular disease. Improved understanding of PCSK9 roles and molecular mechanisms in various vascular diseases will facilitate advances in lipid-lowering therapy and disease prevention.


Asunto(s)
Arterias/enzimología , Hipercolesterolemia/enzimología , Proproteína Convertasa 9/metabolismo , Enfermedades Vasculares/enzimología , Animales , Anticolesterolemiantes/uso terapéutico , Arterias/efectos de los fármacos , Arterias/patología , Regulación Enzimológica de la Expresión Génica , Humanos , Hipercolesterolemia/tratamiento farmacológico , Hipercolesterolemia/genética , Hipercolesterolemia/patología , Mutación , Inhibidores de PCSK9 , Proproteína Convertasa 9/genética , Inhibidores de Serina Proteinasa/uso terapéutico , Transducción de Señal , Enfermedades Vasculares/tratamiento farmacológico , Enfermedades Vasculares/genética , Enfermedades Vasculares/patología
19.
J Cell Physiol ; 236(10): 7159-7175, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33755211

RESUMEN

Atherosclerosis is a significant cardiovascular burden and a leading cause of death worldwide, recognized as a chronic sterile inflammatory disease. Pyroptosis is a novel proinflammatory regulated cell death, characterized by cell swelling, plasma membrane bubbling, and robust release of proinflammatory cytokines (such as interleukin IL-1ß and IL-18). Mounting studies have addressed the crucial contribution of pyroptosis to atherosclerosis and clarified the candidate therapeutic agents targeting pyroptosis for atherosclerosis. Herein, we review the initial characterization of pyroptosis, the detailed mechanisms of pyroptosis, current evidence about pyroptosis and atherosclerosis, and potential therapeutic strategies that target pyroptosis in the development of atherosclerosis.


Asunto(s)
Antiinflamatorios/uso terapéutico , Arterias/efectos de los fármacos , Aterosclerosis/tratamiento farmacológico , Citocinas/metabolismo , Inflamasomas/metabolismo , Mediadores de Inflamación/metabolismo , Piroptosis/efectos de los fármacos , Animales , Arterias/metabolismo , Arterias/patología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Transducción de Señal
20.
Rheumatology (Oxford) ; 60(11): 5052-5059, 2021 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-34117737

RESUMEN

OBJECTIVES: To characterize the effect of ultra-short glucocorticoids followed by Tocilizumab monotherapy on the intima-media thickness (IMT) in GCA. METHODS: Eighteen GCA patients received 500 mg for 3 consecutive days (total of 1500mg) i.v. methylprednisolone on days 0-2, followed by i.v. Tocilizumab (8 mg/kg) on day 3 and thereafter weekly s.c. Tocilizumab injections (162 mg) over 52 weeks. US of temporal (TAs), axillary (AAs) and subclavian (SAs) arteries was performed at baseline, on days 2-3, and at weeks 4, 8, 12, 24 and 52. The largest IMT of all segments and IMT at landmarks of AA/SA were recorded. IMT was scaled by mean normal values and averaged. Each segment was classified according to diagnostic cut-offs. RESULTS: Of the 18 GCA patients, 16 patients had TA and 6 had extracranial large artery involvement. The IMT showed a sharp decline on day 2/3 in the TAs and AAs/SAs. In TAs, this was followed by an increase to baseline levels at week 4 and a subsequent slow decrease, which was paralleled by decreasing symptoms and achievement of clinical remission. The AAs/SAs showed a new signal of vasculitis at week 4 in three patients, with an IMT increase up to week 8. CONCLUSION: Glucocorticoid pulse therapy induced a transient decrease of the IMT in TAs and AAs/SAs. Tocilizumab monotherapy resulted in a slow and steady decrease in IMT of the TAs and a smaller and delayed effect on the AAs/SAs. The data strongly support a remission-inducing effect of Tocilizumab and argue the case for US having an important role in monitoring disease activity in GCA. TRIAL REGISTRATION: ClinicalTrials.gov, www.clinicaltrials.gov, NCT03745586.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Arteritis de Células Gigantes/diagnóstico por imagen , Arteritis de Células Gigantes/tratamiento farmacológico , Glucocorticoides/uso terapéutico , Anciano , Anticuerpos Monoclonales Humanizados/farmacología , Arterias/diagnóstico por imagen , Arterias/efectos de los fármacos , Femenino , Glucocorticoides/farmacología , Humanos , Masculino , Prueba de Estudio Conceptual , Túnica Íntima/diagnóstico por imagen , Túnica Íntima/efectos de los fármacos , Ultrasonografía
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda