Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Brain Behav Immun ; 120: 99-116, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38705494

RESUMEN

INTRODUCTION: Despite improved management of traumatic brain injury (TBI), it still leads to lifelong sequelae and disability, particularly in children. Chronic neuroinflammation (the so-called tertiary phase), in particular, microglia/macrophage and astrocyte reactivity, is among the main mechanisms suspected of playing a role in the generation of lesions associated with TBI. The role of acute neuroinflammation is now well understood, but its persistent effect and impact on the brain, particularly during development, are not. Here, we investigated the long-term effects of pediatric TBI on the brain in a mouse model. METHODS: Pediatric TBI was induced in mice on postnatal day (P) 7 by weight-drop trauma. The time course of neuroinflammation and myelination was examined in the TBI mice. They were also assessed by magnetic resonance, functional ultrasound, and behavioral tests at P45. RESULTS: TBI induced robust neuroinflammation, characterized by acute microglia/macrophage and astrocyte reactivity. The long-term consequences of pediatric TBI studied on P45 involved localized scarring astrogliosis, persistent microgliosis associated with a specific transcriptomic signature, and a long-lasting myelination defect consisting of the loss of myelinated axons, a decreased level of myelin binding protein, and severe thinning of the corpus callosum. These results were confirmed by reduced fractional anisotropy, measured by diffusion tensor imaging, and altered inter- and intra-hemispheric connectivity, measured by functional ultrasound imaging. In addition, adolescent mice with pediatric TBI showed persistent social interaction deficits and signs of anxiety and depressive behaviors. CONCLUSIONS: We show that pediatric TBI induces tertiary neuroinflammatory processes associated with white matter lesions and altered behavior. These results support our model as a model for preclinical studies for tertiary lesions following TBI.

2.
Cells ; 13(8)2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38667299

RESUMEN

It has been known for a long time that epileptic seizures provoke brain neuroinflammation involving the activation of microglial cells. However, the role of these cells in this disease context and the consequences of their inflammatory activation on subsequent neuron network activity remain poorly understood so far. To fill this gap of knowledge and gain a better understanding of the role of microglia in the pathophysiology of epilepsy, we used an established zebrafish Dravet syndrome epilepsy model based on Scn1Lab sodium channel loss-of-function, combined with live microglia and neuronal Ca2+ imaging, local field potential (LFP) recording, and genetic microglia ablation. Data showed that microglial cells in scn1Lab-deficient larvae experiencing epileptiform seizures displayed morphological and biochemical changes characteristic of M1-like pro-inflammatory activation; i.e., reduced branching, amoeboid-like morphology, and marked increase in the number of microglia expressing pro-inflammatory cytokine Il1ß. More importantly, LFP recording, Ca2+ imaging, and swimming behavior analysis showed that microglia-depleted scn1Lab-KD larvae displayed an increase in epileptiform seizure-like neuron activation when compared to that seen in scn1Lab-KD individuals with microglia. These findings strongly suggest that despite microglia activation and the synthesis of pro-inflammatory cytokines, these cells provide neuroprotective activities to epileptic neuronal networks, making these cells a promising therapeutic target in epilepsy.


Asunto(s)
Modelos Animales de Enfermedad , Epilepsias Mioclónicas , Microglía , Neuronas , Pez Cebra , Animales , Microglía/metabolismo , Microglía/patología , Epilepsias Mioclónicas/patología , Epilepsias Mioclónicas/genética , Epilepsias Mioclónicas/metabolismo , Epilepsias Mioclónicas/fisiopatología , Neuronas/metabolismo , Neuronas/patología , Canal de Sodio Activado por Voltaje NAV1.1/genética , Canal de Sodio Activado por Voltaje NAV1.1/metabolismo , Interleucina-1beta/metabolismo , Larva , Calcio/metabolismo , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
5.
Br J Pharmacol ; 180 Suppl 2: S23-S144, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-38123151

RESUMEN

The Concise Guide to PHARMACOLOGY 2023/24 is the sixth in this series of biennial publications. The Concise Guide provides concise overviews, mostly in tabular format, of the key properties of approximately 1800 drug targets, and about 6000 interactions with about 3900 ligands. There is an emphasis on selective pharmacology (where available), plus links to the open access knowledgebase source of drug targets and their ligands (https://www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. Although the Concise Guide constitutes almost 500 pages, the material presented is substantially reduced compared to information and links presented on the website. It provides a permanent, citable, point-in-time record that will survive database updates. The full contents of this section can be found at http://onlinelibrary.wiley.com/doi/bph.16177. G protein-coupled receptors are one of the six major pharmacological targets into which the Guide is divided, with the others being: ion channels, nuclear hormone receptors, catalytic receptors, enzymes and transporters. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. The landscape format of the Concise Guide is designed to facilitate comparison of related targets from material contemporary to mid-2023, and supersedes data presented in the 2021/22, 2019/20, 2017/18, 2015/16 and 2013/14 Concise Guides and previous Guides to Receptors and Channels. It is produced in close conjunction with the Nomenclature and Standards Committee of the International Union of Basic and Clinical Pharmacology (NC-IUPHAR), therefore, providing official IUPHAR classification and nomenclature for human drug targets, where appropriate.


Asunto(s)
Bases de Datos Farmacéuticas , Receptores Acoplados a Proteínas G , Humanos , Ligandos , Canales Iónicos/química , Receptores Citoplasmáticos y Nucleares
8.
Prog Mol Biol Transl Sci ; 196: 43-57, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36813365

RESUMEN

Somatostatin (SRIF) is a neuropeptide that acts as an important regulator of both endocrine and exocrine secretion and modulates neurotransmission in the central nervous system (CNS). SRIF also regulates cell proliferation in normal tissues and tumors. The physiological actions of SRIF are mediated by a family of five G protein-coupled receptors, called somatostatin receptor (SST) SST1, SST2, SST3, SST4, SST5. These five receptors share similar molecular structure and signaling pathways but they display marked differences in their anatomical distribution, subcellular localization and intracellular trafficking. The SST subtypes are widely distributed in the CNS and peripheral nervous system, in many endocrine glands and tumors, particularly of neuroendocrine origin. In this review, we focus on the agonist-dependent internalization and recycling of the different SST subtypes in vivo in the CNS, peripheral organs and tumors. We also discuss the physiological, pathophysiological and potential therapeutic effects of the intracellular trafficking of SST subtypes.


Asunto(s)
Neoplasias , Receptores de Somatostatina , Humanos , Receptores de Somatostatina/química , Receptores de Somatostatina/metabolismo , Encéfalo/metabolismo
9.
J Neural Transm (Vienna) ; 130(3): 281-297, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36335540

RESUMEN

Approximately 15 million babies are born prematurely every year and many will face lifetime motor and/or cognitive deficits. Children born prematurely are at higher risk of developing perinatal brain lesions, especially white matter injuries (WMI). Evidence in humans and rodents demonstrates that systemic inflammation-induced neuroinflammation, including microglial and astrocyte reactivity, is the prominent processes of WMI associated with preterm birth. Thus, a new challenge in the field of perinatal brain injuries is to develop new neuroprotective strategies to target neuroinflammation to prevent WMI. Serotonin (5-HT) and its receptors play an important role in inflammation, and emerging evidence indicates that 5-HT may regulate brain inflammation by the modulation of microglial reactivity and astrocyte functions. The present study is based on a mouse model of WMI induced by intraperitoneal (i.p.) injections of IL-1ß during the first 5 days of life. In this model, certain key lesions of preterm brain injuries can be summarized by (i) systemic inflammation, (ii) pro-inflammatory microglial and astrocyte activation, and (iii) inhibition of oligodendrocyte maturation, leading to hypomyelination. We demonstrate that Htr7 mRNA (coding for the HTR7/5-HT7 receptor) is significantly overexpressed in the anterior cortex of IL-1ß-exposed animals, suggesting it as a potential therapeutic target. LP-211 is a specific high-affinity HTR7 agonist that crosses the blood-brain barrier (BBB). When co-injected with IL-1ß, LP-211 treatment prevented glial reactivity, the down-regulation of myelin-associated proteins, and the apparition of anxiety-like phenotypes. Thus, HTR7 may represent an innovative therapeutic target to protect the developing brain from preterm brain injuries.


Asunto(s)
Lesiones Encefálicas , Nacimiento Prematuro , Sustancia Blanca , Animales , Ratones , Embarazo , Femenino , Niño , Recién Nacido , Humanos , Sustancia Blanca/patología , Roedores , Enfermedades Neuroinflamatorias , Serotonina/metabolismo , Nacimiento Prematuro/metabolismo , Nacimiento Prematuro/patología , Encéfalo/metabolismo , Lesiones Encefálicas/etiología , Lesiones Encefálicas/prevención & control , Inflamación/patología , Microglía/metabolismo
10.
Int J Mol Sci ; 23(9)2022 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-35563258

RESUMEN

Preterm birth (PTB) represents 15 million births every year worldwide and is frequently associated with maternal/fetal infections and inflammation, inducing neuroinflammation. This neuroinflammation is mediated by microglial cells, which are brain-resident macrophages that release cytotoxic molecules that block oligodendrocyte differentiation, leading to hypomyelination. Some preterm survivors can face lifetime motor and/or cognitive disabilities linked to periventricular white matter injuries (PWMIs). There is currently no recommendation concerning the mode of delivery in the case of PTB and its impact on brain development. Many animal models of induced-PTB based on LPS injections exist, but with a low survival rate. There is a lack of information regarding clinically used pharmacological substances to induce PTB and their consequences on brain development. Mifepristone (RU-486) is a drug used clinically to induce preterm labor. This study aims to elaborate and characterize a new model of induced-PTB and PWMIs by the gestational injection of RU-486 and the perinatal injection of pups with IL-1beta. A RU-486 single subcutaneous (s.c.) injection at embryonic day (E)18.5 induced PTB at E19.5 in pregnant OF1 mice. All pups were born alive and were adopted directly after birth. IL-1beta was injected intraperitoneally from postnatal day (P)1 to P5. Animals exposed to both RU-486 and IL-1beta demonstrated microglial reactivity and subsequent PWMIs. In conclusion, the s.c. administration of RU-486 induced labor within 24 h with a high survival rate for pups. In the context of perinatal inflammation, RU-486 labor induction significantly decreases microglial reactivity in vivo but did not prevent subsequent PWMIs.


Asunto(s)
Microglía , Nacimiento Prematuro , Animales , Animales Recién Nacidos , Femenino , Humanos , Inflamación , Lipopolisacáridos/toxicidad , Ratones , Mifepristona/farmacología , Embarazo
11.
Glia ; 70(9): 1699-1719, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35579329

RESUMEN

Preterm infants often show pathologies of the cerebellum, which are associated with impaired motor performance, lower IQ and poor language skills at school ages. Using a mouse model of inflammation-induced encephalopathy of prematurity driven by systemic administration of pro-inflammatory IL-1ß, we sought to uncover causes of cerebellar damage. In this model, IL-1ß is administered between postnatal day (P) 1 to day 5, a timing equivalent to the last trimester for brain development in humans. Structural MRI analysis revealed that systemic IL-1ß treatment induced specific reductions in gray and white matter volumes of the mouse cerebellar lobules I and II (5% false discovery rate [FDR]) from P15 onwards. Preceding these MRI-detectable cerebellar volume changes, we observed damage to oligodendroglia, with reduced proliferation of OLIG2+ cells at P10 and reduced levels of the myelin proteins myelin basic protein (MBP) and myelin-associated glycoprotein (MAG) at P10 and P15. Increased density of IBA1+ cerebellar microglia were observed both at P5 and P45, with evidence for increased microglial proliferation at P5 and P10. Comparison of the transcriptome of microglia isolated from P5 cerebellums and cerebrums revealed significant enrichment of pro-inflammatory markers in microglia from both regions, but cerebellar microglia displayed a unique type I interferon signaling dysregulation. Collectively, these data suggest that perinatal inflammation driven by systemic IL-1ß leads to specific cerebellar volume deficits, which likely reflect oligodendrocyte pathology downstream of microglial activation. Further studies are now required to confirm the potential of protective strategies aimed at preventing sustained type I interferon signaling driven by cerebellar microglia as an important therapeutic target.


Asunto(s)
Enfermedades Cerebelosas , Enfermedades del Prematuro , Inflamación , Interferón Tipo I , Interleucina-1beta , Microglía , Animales , Encefalopatías/inducido químicamente , Encefalopatías/inmunología , Encefalopatías/patología , Enfermedades Cerebelosas/inducido químicamente , Enfermedades Cerebelosas/inmunología , Enfermedades Cerebelosas/patología , Cerebelo/efectos de los fármacos , Cerebelo/inmunología , Cerebelo/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Recién Nacido , Recien Nacido Prematuro , Enfermedades del Prematuro/inducido químicamente , Enfermedades del Prematuro/inmunología , Enfermedades del Prematuro/patología , Inflamación/inducido químicamente , Inflamación/inmunología , Inflamación/patología , Interferón Tipo I/inmunología , Interleucina-1beta/efectos adversos , Interleucina-1beta/farmacología , Microglía/efectos de los fármacos , Microglía/inmunología , Microglía/patología , Embarazo
12.
Ann Neurol ; 91(1): 48-65, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34741343

RESUMEN

OBJECTIVES: In the premature newborn, perinatal inflammation mediated by microglia contributes significantly to neurodevelopmental injuries including white matter injury (WMI). Brain inflammation alters development through neuroinflammatory processes mediated by activation of homeostatic microglia toward a pro-inflammatory and neurotoxic phenotype. Investigating immune regulators of microglial activation is crucial to find effective strategies to prevent and treat WMI. METHODS: Ex vivo microglial cultures and a mouse model of WMI induced by perinatal inflammation (interleukin-1-beta [IL-1ß] and postnatal days 1-5) were used to uncover and elucidate the role of microRNA-146b-5p in microglial activation and WMI. RESULTS: A specific reduction in vivo in microglia of Dicer, a protein required for microRNAs maturation, reduces pro-inflammatory activation of microglia and prevents hypomyelination in our model of WMI. Microglial miRNome analysis in the WMI model identified miRNA-146b-5p as a candidate modulator of microglial activation. Ex vivo microglial cell culture treated with the pro-inflammatory stimulus lipopolysaccharide (LPS) led to overexpression of immunomodulatory miRNA-146b-5p but its drastic reduction in the microglial extracellular vesicles (EVs). To increase miRNA-146b-5p expression, we used a 3DNA nanocarrier to deliver synthetic miRNA-146b-5p specifically to microglia. Enhancing microglial miRNA-146b-5p overexpression significantly decreased LPS-induced activation, downregulated IRAK1, and restored miRNA-146b-5p levels in EVs. In our WMI model, 3DNA miRNA-146b-5p treatment significantly prevented microglial activation, hypomyelination, and cognitive defect induced by perinatal inflammation. INTERPRETATIONS: These findings support that miRNA-146b-5p is a major regulator of microglia phenotype and could be targeted to reduce the incidence and the severity of perinatal brain injuries and their long-term consequences. ANN NEUROL 2022;91:48-65.


Asunto(s)
Encéfalo/patología , MicroARNs/metabolismo , Microglía/patología , Sustancia Blanca/patología , Animales , Ratones , Neurogénesis/fisiología
13.
J Exp Orthop ; 8(1): 116, 2021 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-34897564

RESUMEN

PURPOSE: The revision of any total knee replacement is carried out in a significant number of cases, due to the excessive internal rotation of the tibial component. The goal was to develop a personalized method, using only the geometric parameters of the tibia, without the femoral guidelines, to calculate the postoperative rotational position of tibial component malrotation within a tolerable error threshold in every case. METHODS: Preoperative CT scans of eighty-five osteoarthritic knees were examined by three independent medical doctors twice over 7 weeks. The geometric centre of the tibia was produced by the ellipse annotation drawn 8 mm below the tibial plateau, the sagittal and frontal axes of the ellipse were transposed to the slice of the tibial tuberosity. With the usage of several guide lines, a right triangle was drawn within which the personalized Berger angle was calculated. RESULTS: A very good intra-observer (0.89-0.925) and inter-observer (0.874) intra-class correlation coefficient (ICC) was achieved. Even if the average of the personalized Berger values were similar to the original 18° (18.32° in our case), only 70.6% of the patients are between the clinically tolerable thresholds (12.2° and 23.8°). CONCLUSION: The method, measured on the preoperative CT scans, is capable of calculating the required correction during the planning of revision arthroplasties which are necessary due to the tibial component malrotation. The personalized Berger angle isn't altered during arthroplasty, this way it determines which one of the anterior reference points of the tibia (medial 1/3 or the tip of the tibial tuberosity, medial border or 1/6 or 1/3 or the centre of the patellar tendon) can be used during the positioning of the tibial component. LEVEL OF EVIDENCE: Level II, Diagnostic Study (Methodological Study).

14.
Br J Pharmacol ; 178 Suppl 1: S27-S156, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34529832

RESUMEN

The Concise Guide to PHARMACOLOGY 2021/22 is the fifth in this series of biennial publications. The Concise Guide provides concise overviews, mostly in tabular format, of the key properties of nearly 1900 human drug targets with an emphasis on selective pharmacology (where available), plus links to the open access knowledgebase source of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. Although the Concise Guide constitutes over 500 pages, the material presented is substantially reduced compared to information and links presented on the website. It provides a permanent, citable, point-in-time record that will survive database updates. The full contents of this section can be found at http://onlinelibrary.wiley.com/doi/bph.15538. G protein-coupled receptors are one of the six major pharmacological targets into which the Guide is divided, with the others being: ion channels, nuclear hormone receptors, catalytic receptors, enzymes and transporters. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. The landscape format of the Concise Guide is designed to facilitate comparison of related targets from material contemporary to mid-2021, and supersedes data presented in the 2019/20, 2017/18, 2015/16 and 2013/14 Concise Guides and previous Guides to Receptors and Channels. It is produced in close conjunction with the Nomenclature and Standards Committee of the International Union of Basic and Clinical Pharmacology (NC-IUPHAR), therefore, providing official IUPHAR classification and nomenclature for human drug targets, where appropriate.


Asunto(s)
Bases de Datos Farmacéuticas , Farmacología , Humanos , Canales Iónicos , Ligandos , Receptores Citoplasmáticos y Nucleares , Receptores Acoplados a Proteínas G
15.
Cell Death Dis ; 12(2): 166, 2021 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-33558485

RESUMEN

A leading cause of preterm birth is the exposure to systemic inflammation (maternal/fetal infection), which leads to neuroinflammation and white matter injury (WMI). A wide range of cytokines and chemokines are expressed and upregulated in oligodendrocytes (OLs) in response to inflammation and numerous reports show that OLs express several receptors for immune related molecules, which enable them to sense inflammation and to react. However, the role of OL immune response in WMI is unclear. Here, we focus our study on toll-like receptor-3 (TLR3) that is activated by double-strand RNA (dsRNA) and promotes neuroinflammation. Despite its importance, its expression and role in OLs remain unclear. We used an in vivo mouse model, which mimics inflammation-mediated WMI of preterm born infants consisting of intraperitoneal injection of IL-1ß from P1 to P5. In the IL-1ß-treated animals, we observed the upregulation of Tlr3, IL-1ß, IFN-ß, Ccl2, and Cxcl10 in both PDGFRα+ and O4+ sorted cells. This upregulation was higher in O4+ immature OLs (immOLs) as compared to PDGFRα+ OL precursor cells (OPCs), suggesting a different sensitivity to neuroinflammation. These observations were confirmed in OL primary cultures: cells treated with TLR3 agonist Poly(I:C) during differentiation showed a stronger upregulation of Ccl2 and Cxcl10 compared to cells treated during proliferation and led to decreased expression of myelin genes. Finally, OLs were able to modulate microglia phenotype and function depending on their maturation state as assessed by qPCR using validated markers for immunomodulatory, proinflammatory, and anti-inflammatory phenotypes and by phagocytosis and morphological analysis. These results show that during inflammation the response of OLs can play an autonomous role in blocking their own differentiation: in addition, the immune activation of OLs may play an important role in shaping the response of microglia during inflammation.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Encefalitis/metabolismo , Leucoencefalopatías/metabolismo , Oligodendroglía/metabolismo , Receptor Toll-Like 3/metabolismo , Sustancia Blanca/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalitis/genética , Encefalitis/inmunología , Encefalitis/patología , Femenino , Mediadores de Inflamación/metabolismo , Leucoencefalopatías/genética , Leucoencefalopatías/inmunología , Leucoencefalopatías/patología , Masculino , Ratones , Microglía/inmunología , Microglía/metabolismo , Microglía/patología , Oligodendroglía/efectos de los fármacos , Oligodendroglía/inmunología , Oligodendroglía/patología , Poli I-C/farmacología , Embarazo , Nacimiento Prematuro , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Receptor Toll-Like 3/agonistas , Sustancia Blanca/efectos de los fármacos , Sustancia Blanca/inmunología , Sustancia Blanca/patología
16.
Cell Rep ; 31(2): 107506, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32294449

RESUMEN

A distinctive feature of neocortical development is the highly coordinated production of different progenitor cell subtypes, which are critical for ensuring adequate neurogenic outcome and the development of normal neocortical size. To further understand the mechanisms that underlie neocortical growth, we focused our studies on the microcephaly gene Mcph1, and we report here that Mcph1 (1) exerts its functions in rapidly dividing apical radial glial cells (aRGCs) during mouse neocortical development stages that precede indirect neurogenesis; (2) is expressed at mitochondria; and (3) controls the proper proliferation and survival of RGCs, potentially through crosstalk with cellular metabolic pathways involving the stimulation of mitochondrial activity via VDAC1/GRP75 and AKT/HK2/VDAC1 and glutaminolysis via ATF4/PCK2. We currently report the description of a MCPH-gene implication in the interplay between bioenergetic pathways and neocortical growth, thus pointing to alterations of cellular metabolic pathways, in particular glutaminolysis, as a possible cause of microcephalic pathogenesis.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas del Citoesqueleto/genética , Microcefalia/genética , Microcefalia/metabolismo , Animales , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular/genética , Proliferación Celular/genética , Supervivencia Celular/genética , Proteínas del Citoesqueleto/metabolismo , Femenino , Células HEK293 , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Microcefalia/fisiopatología , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Mutación , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/genética , Neuroglía/metabolismo , Neuronas/metabolismo , Canal Aniónico 1 Dependiente del Voltaje/genética , Canal Aniónico 1 Dependiente del Voltaje/metabolismo
17.
Brain ; 142(12): 3806-3833, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31665242

RESUMEN

Microglia of the developing brain have unique functional properties but how their activation states are regulated is poorly understood. Inflammatory activation of microglia in the still-developing brain of preterm-born infants is associated with permanent neurological sequelae in 9 million infants every year. Investigating the regulators of microglial activation in the developing brain across models of neuroinflammation-mediated injury (mouse, zebrafish) and primary human and mouse microglia we found using analysis of genes and proteins that a reduction in Wnt/ß-catenin signalling is necessary and sufficient to drive a microglial phenotype causing hypomyelination. We validated in a cohort of preterm-born infants that genomic variation in the Wnt pathway is associated with the levels of connectivity found in their brains. Using a Wnt agonist delivered by a blood-brain barrier penetrant microglia-specific targeting nanocarrier we prevented in our animal model the pro-inflammatory microglial activation, white matter injury and behavioural deficits. Collectively, these data validate that the Wnt pathway regulates microglial activation, is critical in the evolution of an important form of human brain injury and is a viable therapeutic target.


Asunto(s)
Encéfalo/metabolismo , Inflamación/metabolismo , Microglía/metabolismo , Vía de Señalización Wnt/fisiología , Animales , Animales Modificados Genéticamente , Barrera Hematoencefálica/metabolismo , Células Cultivadas , Biología Computacional , Humanos , Ratones , Pez Cebra
18.
Quant Imaging Med Surg ; 9(1): 115-126, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30788253

RESUMEN

Education and training of morphology for medical students, and professionals specializing in pediatric cardiology and surgery has traditionally been based on hands-on encounter with congenitally malformed cardiac specimens. Large international archives are no longer widely available due to stricter data protection rules, a reduced number of autopsies, attrition rate of existing specimens, and most importantly due to a higher survival rate of patients. Our Cardiac Archive houses about 400 cardiac specimens with congenital heart disease. The collection spans almost 60 years and thus goes back to pre-surgical era. Unfortunately, attrition rate due to desiccation has led to an increased natural decay in recent years. The present multi-institutional project focuses on saving the collection by digitization. Specimens are scanned by high-resolution micro-CT/MRI. Virtual 3D-models are segmented and a comprehensive database is built. We now report an initial feasibility study with six test specimens that provided promising results, however, adequate presentation of the intracardiac anatomy, including septa and cardiac valves requires further refinements. Computer assisted design methods are necessary to overcome consequences of pathological examination, shrinkage and/or distortion of the specimens. For a next step, we anticipate an expandable web-based virtual museum with interactive reference and training tools. Web access for professional third parties will be provided by registration/subscription. In a future phase, segmental wall motion data could be added to virtual models. 3D-printed models may replace actual specimens and serve as hands-on surgical training to elucidate complex morphologies, promote surgical emulation, and extract more accurate procedural knowledge based on such a collection.

19.
Front Cell Neurosci ; 12: 450, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30555302

RESUMEN

Our aim was to examine the dynamics of the muscarinic m2 receptor (m2R), a G-protein coupled receptor (GPCR), after agonist activation in living hippocampal neurons, and especially clathrin dependency endocytosis. We have previously shown that the m2R undergoes agonist-induced internalization in vivo. However, the nature of the endocytotic pathway used by m2R after activation is still unknown in living neurons. Using live cell imaging and quantitative analyses, we have monitored the effect of stimulation on the fate of the membrane-bound m2R and on its redistribution in intraneuronal compartments. Shortly (6 min) after activation, m2R is internalized into clathrin immunopositive structures. Furthermore, after clathrin-dependent endocytosis, m2R associates with early and late endosomes and with subcellular organelles involved in degradation. Together, these results provide, for the first time, a description of m2R trafficking in living neurons and prove that m2R undergoes clathrin-dependent endocytosis before being degraded.

20.
Pharmacol Rev ; 70(4): 763-835, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30232095

RESUMEN

Somatostatin, also known as somatotropin-release inhibitory factor, is a cyclopeptide that exerts potent inhibitory actions on hormone secretion and neuronal excitability. Its physiologic functions are mediated by five G protein-coupled receptors (GPCRs) called somatostatin receptor (SST)1-5. These five receptors share common structural features and signaling mechanisms but differ in their cellular and subcellular localization and mode of regulation. SST2 and SST5 receptors have evolved as primary targets for pharmacological treatment of pituitary adenomas and neuroendocrine tumors. In addition, SST2 is a prototypical GPCR for the development of peptide-based radiopharmaceuticals for diagnostic and therapeutic interventions. This review article summarizes findings published in the last 25 years on the physiology, pharmacology, and clinical applications related to SSTs. We also discuss potential future developments and propose a new nomenclature.


Asunto(s)
Receptores de Somatostatina/metabolismo , Animales , Regulación de la Expresión Génica , Humanos , Ligandos , Conformación Proteica , Transporte de Proteínas , Receptores de Somatostatina/química , Receptores de Somatostatina/genética , Receptores de Somatostatina/fisiología , Transducción de Señal , Terminología como Asunto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA