Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Nature ; 605(7908): 152-159, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35477759

RESUMEN

Atherosclerotic plaques develop in the inner intimal layer of arteries and can cause heart attacks and strokes1. As plaques lack innervation, the effects of neuronal control on atherosclerosis remain unclear. However, the immune system responds to plaques by forming leukocyte infiltrates in the outer connective tissue coat of arteries (the adventitia)2-6. Here, because the peripheral nervous system uses the adventitia as its principal conduit to reach distant targets7-9, we postulated that the peripheral nervous system may directly interact with diseased arteries. Unexpectedly, widespread neuroimmune cardiovascular interfaces (NICIs) arose in mouse and human atherosclerosis-diseased adventitia segments showed expanded axon networks, including growth cones at axon endings near immune cells and media smooth muscle cells. Mouse NICIs established a structural artery-brain circuit (ABC): abdominal adventitia nociceptive afferents10-14 entered the central nervous system through spinal cord T6-T13 dorsal root ganglia and were traced to higher brain regions, including the parabrachial and central amygdala neurons; and sympathetic efferent neurons projected from medullary and hypothalamic neurons to the adventitia through spinal intermediolateral neurons and both coeliac and sympathetic chain ganglia. Moreover, ABC peripheral nervous system components were activated: splenic sympathetic and coeliac vagus nerve activities increased in parallel to disease progression, whereas coeliac ganglionectomy led to the disintegration of adventitial NICIs, reduced disease progression and enhanced plaque stability. Thus, the peripheral nervous system uses NICIs to assemble a structural ABC, and therapeutic intervention in the ABC attenuates atherosclerosis.


Asunto(s)
Aterosclerosis , Placa Aterosclerótica , Animales , Aterosclerosis/prevención & control , Progresión de la Enfermedad , Ganglios Espinales , Ganglios Simpáticos , Ratones , Neuronas/fisiología , Placa Aterosclerótica/prevención & control
2.
Immunity ; 47(5): 959-973.e9, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29150241

RESUMEN

Aortic aneurysms are life-threatening conditions with effective treatments mainly limited to emergency surgery or trans-arterial endovascular stent grafts, thus calling for the identification of specific molecular targets. Genetic studies have highlighted controversial roles of transforming growth factor ß (TGF-ß) signaling in aneurysm development. Here, we report on aneurysms developing in adult mice after smooth muscle cell (SMC)-specific inactivation of Smad4, an intracellular transducer of TGF-ß. The results revealed that Smad4 inhibition activated interleukin-1ß (IL-1ß) in SMCs. This danger signal later recruited innate immunity in the adventitia through chemokine (C-C motif) ligand 2 (CCL2) and modified the mechanical properties of the aortic wall, thus favoring vessel dilation. SMC-specific Smad4 deletion in Il1r1- or Ccr2-null mice resulted in milder aortic pathology. A chronic treatment with anti-IL-1ß antibody effectively hampered aneurysm development. These findings identify a mechanistic target for controlling the progression of aneurysms with compromised TGF-ß signaling, such as those driven by SMAD4 mutations.


Asunto(s)
Aneurisma de la Aorta/prevención & control , Interleucina-1beta/antagonistas & inhibidores , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/fisiología , Animales , Células Cultivadas , Quimiocina CCL2/antagonistas & inhibidores , Interleucina-1beta/biosíntesis , Ratones , Miocitos del Músculo Liso/inmunología , FN-kappa B/fisiología , Receptores CCR2/antagonistas & inhibidores , Proteína Smad4/fisiología , Tamoxifeno/farmacología
3.
Hypertension ; 81(3): 636-647, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38174566

RESUMEN

BACKGROUND: Hypertension is one of the main risk factors for dementia and cognitive impairment. METHODS: We used the model of transverse aortic constriction to induce chronic pressure overload in mice. We characterized brain injury by advanced translational applications of magnetic resonance imaging. In parallel, we analyzed peripheral target organ damage induced by chronic pressure overload by ultrasonography. Microscopical characterization of brain vasculature was performed as well, together with the analysis of immune and inflammatory markers. RESULTS: We identified a specific structural, microstructural, and functional brain injury. In particular, we highlighted a regional enlargement of the hypothalamus, microstructural damage in the white matter of the fimbria, and a reduction of the cerebral blood flow. A parallel analysis performed by confocal microscopy revealed a correspondent tissue damage evidenced by a reduction of cerebral capillary density, paired with loss of pericyte coverage. We assessed cognitive impairment and cardiac damage induced by hypertension to perform correlation analyses with the brain injury severity. At the mechanistic level, we found that CD8+T cells, producing interferon-γ, infiltrated the brain of hypertensive mice. By neutralizing this proinflammatory cytokine, we obtained a rescue of the phenotype, demonstrating their crucial role in establishing the microvascular damage. CONCLUSIONS: Overall, we have used translational tools to comprehensively characterize brain injury in a mouse model of hypertension induced by chronic pressure overload. We have identified early cerebrovascular damage in hypertensive mice, sustained by CD8+IFN-γ+T lymphocytes, which fuel neuroinflammation to establish the injury of brain capillaries.


Asunto(s)
Lesiones Encefálicas , Hipertensión , Ratones , Animales , Enfermedades Neuroinflamatorias , Encéfalo/patología , Imagen por Resonancia Magnética , Lesiones Encefálicas/patología
4.
Cardiovasc Res ; 119(5): 1234-1249, 2023 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-36536484

RESUMEN

AIMS: Dysregulated immune response contributes to inefficiency of treatment strategies to control hypertension and reduce the risk of end-organ damage. Uncovering the immune pathways driving the transition from the onset of hypertensive stimulus to the manifestation of multi-organ dysfunction are much-needed insights for immune targeted therapy. METHODS AND RESULTS: To aid visualization of cellular events orchestrating multi-organ pathogenesis, we modelled hypertensive cardiovascular remodelling in zebrafish. Zebrafish larvae exposed to ion-poor environment exhibited rapid angiotensinogen up-regulation, followed by manifestation of arterial hypertension and cardiac remodelling that recapitulates key characteristics of incipient heart failure with preserved ejection fraction. In the brain, time-lapse imaging revealed the occurrence of cerebrovascular regression through endothelial retraction and migration in response to the ion-poor treatment. This phenomenon is associated with macrophage/microglia-endothelial contacts and endothelial junctional retraction. Cytokine and transcriptomic profiling identified systemic up-regulation of interferon-γ and interleukin 1ß and revealed altered macrophage/microglia transcriptional programme characterized by suppression of innate immunity and vasculo/neuroprotective gene expression. Both zebrafish and a murine model of pressure overload-induced brain damage demonstrated that the brain pathology and macrophage/microglia phenotypic alteration are dependent on interferon-γ signalling. In zebrafish, interferon-γ receptor 1 mutation prevents cerebrovascular remodelling and dysregulation of macrophage/microglia transcriptomic profile. Supplementation of bone morphogenetic protein 5 identified from the transcriptomic approach as a down-regulated gene in ion-poor-treated macrophages/microglia that is rescued by interferon-γ blockage, mitigated cerebral microvessel loss. In mice subjected to transverse aortic constriction-induced pressure overload, typically developing cerebrovascular injury, neuroinflammation, and cognitive dysfunction, interferon-γ neutralization protected them from blood-brain barrier disruption, cerebrovascular rarefaction, and cognitive decline. CONCLUSIONS: These findings uncover cellular and molecular players of an immune pathway communicating hypertensive stimulus to structural and functional remodelling of the brain and identify anti-interferon-γ treatment as a promising intervention strategy capable of preventing pressure overload-induced damage of the cerebrovascular and nervous systems.


Asunto(s)
Disfunción Cognitiva , Hipertensión , Ratones , Animales , Pez Cebra/metabolismo , Modelos Animales de Enfermedad , Macrófagos/metabolismo , Interferón gamma/metabolismo
5.
Nat Cardiovasc Res ; 2(12): 1277-1290, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38344689

RESUMEN

After myocardial infarction (MI), emergency hematopoiesis produces inflammatory myeloid cells that accelerate atherosclerosis and promote heart failure. Since the balance between glycolysis and mitochondrial metabolism regulates hematopoietic stem cell homeostasis, metabolic cues may influence emergency myelopoiesis. Here, we show in humans and female mice that hematopoietic progenitor cells increase fatty acid metabolism after MI. Blockade of fatty acid oxidation by deleting carnitine palmitoyltransferase (Cpt1A) in hematopoietic cells of Vav1Cre/+Cpt1Afl/fl mice limited hematopoietic progenitor proliferation and myeloid cell expansion after MI. We also observed reduced bone marrow adiposity in humans, pigs and mice following MI. Inhibiting lipolysis in adipocytes using AdipoqCreERT2Atglfl/fl mice or local depletion of bone marrow adipocytes in AdipoqCreERT2iDTR mice also curbed emergency hematopoiesis. Furthermore, systemic and regional sympathectomy prevented bone marrow adipocyte shrinkage after MI. These data establish a critical role for fatty acid metabolism in post-MI emergency hematopoiesis.

6.
Rev Esp Cardiol (Engl Ed) ; 70(1): 16-24, 2017 Jan.
Artículo en Inglés, Español | MEDLINE | ID: mdl-27422446

RESUMEN

INTRODUCTION AND OBJECTIVES: Cardiovascular diseases, including cardiomyopathy, are the major complications in diabetes. A deeper understanding of the molecular mechanisms leading to cardiomyopathy is critical for developing novel therapies. We proposed phosphoinositide3-kinase gamma (PI3Kγ) as a molecular target against diabetic cardiomyopathy, given the role of PI3Kγ in cardiac remodeling to pressure overload. Given the availability of a pharmacological inhibitor of this molecular target GE21, we tested the validity of our hypothesis by inducing diabetes in mice with genetic ablation of PI3Kγ or knock-in for a catalytically inactive PI3Kγ. METHODS: Mice were made diabetic by streptozotocin. Cardiac function was assessed by serial echocardiographic analyses, while fibrosis and inflammation were evaluated by histological analysis. RESULTS: Diabetes induced cardiac dysfunction in wild-type mice. Systolic dysfunction was completely prevented, and diastolic dysfunction was partially blocked, in both PI3Kγ knock-out and kinase-dead mice. Cardiac dysfunction was similarly rescued by administration of the PI3Kγ inhibitor GE21 in a dose-dependent manner. These actions of genetic and pharmacological PI3Kγ inhibition were associated with a decrease in inflammation and fibrosis in diabetic hearts. CONCLUSIONS: Our study demonstrates a fundamental role of PI3Kγ in diabetic cardiomyopathy in mice and the beneficial effect of pharmacological PI3Kγ inhibition, highlighting its potential as a promising strategy for clinical treatment of cardiac complications of diabetic patients.


Asunto(s)
Cardiomiopatías Diabéticas/tratamiento farmacológico , Inhibidores de las Quinasa Fosfoinosítidos-3 , Animales , Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Cardiomiopatías Diabéticas/diagnóstico , Cardiomiopatías Diabéticas/enzimología , Cardiomiopatías Diabéticas/fisiopatología , Modelos Animales de Enfermedad , Ecocardiografía , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/enzimología , Miocardio/patología
8.
Nat Commun ; 7: 13035, 2016 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-27676657

RESUMEN

The crucial role of the immune system in hypertension is now widely recognized. We previously reported that hypertensive challenges couple the nervous drive with immune system activation, but the physiological and molecular mechanisms of this connection are unknown. Here, we show that hypertensive challenges activate splenic sympathetic nerve discharge to prime immune response. More specifically, a vagus-splenic nerve drive, mediated by nicotinic cholinergic receptors, links the brain and spleen. The sympathetic discharge induced by hypertensive stimuli was absent in both coeliac vagotomized mice and in mice lacking α7nAChR, a receptor typically expressed by peripheral ganglionic neurons. This cholinergic-sympathetic pathway is necessary for T cell activation and egression on hypertensive challenges. In addition, we show that selectively thermoablating the splenic nerve prevents T cell egression and protects against hypertension. This novel experimental procedure for selective splenic denervation suggests new clinical strategies for resistant hypertension.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA