Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 127
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 182(3): 734-743.e5, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32643603

RESUMEN

COVID-19, caused by SARS-CoV-2, is a virulent pneumonia, with >4,000,000 confirmed cases worldwide and >290,000 deaths as of May 15, 2020. It is critical that vaccines and therapeutics be developed very rapidly. Mice, the ideal animal for assessing such interventions, are resistant to SARS-CoV-2. Here, we overcome this difficulty by exogenous delivery of human ACE2 with a replication-deficient adenovirus (Ad5-hACE2). Ad5-hACE2-sensitized mice developed pneumonia characterized by weight loss, severe pulmonary pathology, and high-titer virus replication in lungs. Type I interferon, T cells, and, most importantly, signal transducer and activator of transcription 1 (STAT1) are critical for virus clearance and disease resolution in these mice. Ad5-hACE2-transduced mice enabled rapid assessments of a vaccine candidate, of human convalescent plasma, and of two antiviral therapies (poly I:C and remdesivir). In summary, we describe a murine model of broad and immediate utility to investigate COVID-19 pathogenesis and to evaluate new therapies and vaccines.


Asunto(s)
Betacoronavirus/inmunología , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/prevención & control , Modelos Animales de Enfermedad , Pandemias/prevención & control , Neumonía Viral/patología , Neumonía Viral/prevención & control , Vacunación , Enzima Convertidora de Angiotensina 2 , Animales , COVID-19 , Chlorocebus aethiops , Infecciones por Coronavirus/virología , Evaluación Preclínica de Medicamentos/métodos , Femenino , Humanos , Interferón gamma/genética , Interferón gamma/metabolismo , Pulmón/patología , Pulmón/virología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/virología , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , SARS-CoV-2 , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Organismos Libres de Patógenos Específicos , Transducción Genética , Células Vero , Carga Viral , Replicación Viral
2.
Proc Natl Acad Sci U S A ; 120(4): e2202820120, 2023 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-36652473

RESUMEN

Human coronavirus 229E (HCoV-229E) and NL63 (HCoV-NL63) are endemic causes of upper respiratory infections such as the "common cold" but may occasionally cause severe lower respiratory tract disease in the elderly and immunocompromised patients. There are no approved antiviral drugs or vaccines for these common cold coronaviruses (CCCoV). The recent emergence of COVID-19 and the possible cross-reactive antibody and T cell responses between these CCCoV and SARS-CoV-2 emphasize the need to develop experimental animal models for CCCoV. Mice are an ideal experimental animal model for such studies, but are resistant to HCoV-229E and HCoV-NL63 infections. Here, we generated 229E and NL63 mouse models by exogenous delivery of their receptors, human hAPN and hACE2 using replication-deficient adenoviruses (Ad5-hAPN and Ad5-hACE2), respectively. Ad5-hAPN- and Ad5-hACE2-sensitized IFNAR-/- and STAT1-/- mice developed pneumonia characterized by inflammatory cell infiltration with virus clearance occurring 7 d post infection. Ad5-hAPN- and Ad5-hACE2-sensitized mice generated virus-specific T cells and neutralizing antibodies after 229E or NL63 infection, respectively. Remdesivir and a vaccine candidate targeting spike protein of 229E and NL63 accelerated viral clearance of virus in these mice. 229E- and NL63-infected mice were partially protected from SARS-CoV-2 infection, likely mediated by cross-reactive T cell responses. Ad5-hAPN- and Ad5-hACE2-transduced mice are useful for studying pathogenesis and immune responses induced by HCoV-229E and HCoV-NL63 infections and for validation of broadly protective vaccines, antibodies, and therapeutics against human respiratory coronaviruses including SARS-CoV-2.


Asunto(s)
COVID-19 , Resfriado Común , Coronavirus Humano 229E , Coronavirus Humano NL63 , Humanos , Animales , Ratones , Anciano , SARS-CoV-2 , Protección Cruzada
3.
Genome Res ; 32(2): 228-241, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35064006

RESUMEN

The pathogenesis of COVID-19 is still elusive, which impedes disease progression prediction, differential diagnosis, and targeted therapy. Plasma cell-free RNAs (cfRNAs) carry unique information from human tissue and thus could point to resourceful solutions for pathogenesis and host-pathogen interactions. Here, we performed a comparative analysis of cfRNA profiles between COVID-19 patients and healthy donors using serial plasma. Analyses of the cfRNA landscape, potential gene regulatory mechanisms, dynamic changes in tRNA pools upon infection, and microbial communities were performed. A total of 380 cfRNA molecules were up-regulated in all COVID-19 patients, of which seven could serve as potential biomarkers (AUC > 0.85) with great sensitivity and specificity. Antiviral (NFKB1A, IFITM3, and IFI27) and neutrophil activation (S100A8, CD68, and CD63)-related genes exhibited decreased expression levels during treatment in COVID-19 patients, which is in accordance with the dynamically enhanced inflammatory response in COVID-19 patients. Noncoding RNAs, including some microRNAs (let 7 family) and long noncoding RNAs (GJA9-MYCBP) targeting interleukin (IL6/IL6R), were differentially expressed between COVID-19 patients and healthy donors, which accounts for the potential core mechanism of cytokine storm syndromes; the tRNA pools change significantly between the COVID-19 and healthy group, leading to the accumulation of SARS-CoV-2 biased codons, which facilitate SARS-CoV-2 replication. Finally, several pneumonia-related microorganisms were detected in the plasma of COVID-19 patients, raising the possibility of simultaneously monitoring immune response regulation and microbial communities using cfRNA analysis. This study fills the knowledge gap in the plasma cfRNA landscape of COVID-19 patients and offers insight into the potential mechanisms of cfRNAs to explain COVID-19 pathogenesis.


Asunto(s)
COVID-19 , Ácidos Nucleicos Libres de Células , ARN/sangre , COVID-19/sangre , COVID-19/genética , Ácidos Nucleicos Libres de Células/sangre , Síndrome de Liberación de Citoquinas , Humanos , SARS-CoV-2
4.
Nature ; 568(7752): 336-343, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30996318

RESUMEN

The brains of humans and other mammals are highly vulnerable to interruptions in blood flow and decreases in oxygen levels. Here we describe the restoration and maintenance of microcirculation and molecular and cellular functions of the intact pig brain under ex vivo normothermic conditions up to four hours post-mortem. We have developed an extracorporeal pulsatile-perfusion system and a haemoglobin-based, acellular, non-coagulative, echogenic, and cytoprotective perfusate that promotes recovery from anoxia, reduces reperfusion injury, prevents oedema, and metabolically supports the energy requirements of the brain. With this system, we observed preservation of cytoarchitecture; attenuation of cell death; and restoration of vascular dilatory and glial inflammatory responses, spontaneous synaptic activity, and active cerebral metabolism in the absence of global electrocorticographic activity. These findings demonstrate that under appropriate conditions the isolated, intact large mammalian brain possesses an underappreciated capacity for restoration of microcirculation and molecular and cellular activity after a prolonged post-mortem interval.


Asunto(s)
Autopsia , Encéfalo/irrigación sanguínea , Encéfalo/citología , Circulación Cerebrovascular , Microcirculación , Porcinos , Animales , Encéfalo/metabolismo , Encéfalo/patología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Caspasa 3/metabolismo , Supervivencia Celular , Arterias Cerebrales/fisiología , Modelos Animales de Enfermedad , Hipoxia Encefálica/metabolismo , Hipoxia Encefálica/patología , Inflamación/metabolismo , Inflamación/patología , Neuroglía/citología , Neuronas/citología , Neuronas/metabolismo , Neuronas/patología , Perfusión , Daño por Reperfusión/prevención & control , Porcinos/sangre , Sinapsis/metabolismo , Sinapsis/patología , Factores de Tiempo , Vasodilatación
5.
Proc Natl Acad Sci U S A ; 118(51)2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34911761

RESUMEN

Arterial remodeling is an important adaptive mechanism that maintains normal fluid shear stress in a variety of physiologic and pathologic conditions. Inward remodeling, a process that leads to reduction in arterial diameter, plays a critical role in progression of such common diseases as hypertension and atherosclerosis. Yet, despite its pathogenic importance, molecular mechanisms controlling inward remodeling remain undefined. Mitogen-activated protein kinases (MAPKs) perform a number of functions ranging from control of proliferation to migration and cell-fate transitions. While the MAPK ERK1/2 signaling pathway has been extensively examined in the endothelium, less is known about the role of the MEKK3/ERK5 pathway in vascular remodeling. To better define the role played by this signaling cascade, we studied the effect of endothelial-specific deletion of its key upstream MAP3K, MEKK3, in adult mice. The gene's deletion resulted in a gradual inward remodeling of both pulmonary and systematic arteries, leading to spontaneous hypertension in both vascular circuits and accelerated progression of atherosclerosis in hyperlipidemic mice. Molecular analysis revealed activation of TGFß-signaling both in vitro and in vivo. Endothelial-specific TGFßR1 knockout prevented inward arterial remodeling in MEKK3 endothelial knockout mice. These data point to the unexpected participation of endothelial MEKK3 in regulation of TGFßR1-Smad2/3 signaling and inward arterial remodeling in artery diseases.


Asunto(s)
Hipertensión Pulmonar/patología , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , MAP Quinasa Quinasa Quinasa 3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Remodelación Vascular/fisiología , Animales , Eliminación de Gen , Regulación de la Expresión Génica/efectos de los fármacos , Genotipo , Miembro Posterior/irrigación sanguínea , Células Endoteliales de la Vena Umbilical Humana , Humanos , Hipertensión Pulmonar/metabolismo , Isquemia , Quinasa 1 de Quinasa de Quinasa MAP/genética , MAP Quinasa Quinasa Quinasa 3/genética , Ratones , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Moduladores Selectivos de los Receptores de Estrógeno/toxicidad , Transducción de Señal , Tamoxifeno/toxicidad , Factor de Crecimiento Transformador beta/genética
6.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34504019

RESUMEN

Endothelial cell (EC) sensing of wall fluid shear stress (FSS) from blood flow governs vessel remodeling to maintain FSS at a specific magnitude or set point in healthy vessels. Low FSS triggers inward remodeling to restore normal FSS but the regulatory mechanisms are unknown. In this paper, we describe the signaling network that governs inward artery remodeling. FSS induces Smad2/3 phosphorylation through the type I transforming growth factor (TGF)-ß family receptor Alk5 and the transmembrane protein Neuropilin-1, which together increase sensitivity to circulating bone morphogenetic protein (BMP)-9. Smad2/3 nuclear translocation and target gene expression but not phosphorylation are maximal at low FSS and suppressed at physiological high shear. Reducing flow by carotid ligation in rodents increases Smad2/3 nuclear localization, while the resultant inward remodeling is blocked by the EC-specific deletion of Alk5. The flow-activated MEKK3/Klf2 pathway mediates the suppression of Smad2/3 nuclear translocation at high FSS, mainly through the cyclin-dependent kinase (CDK)-2-dependent phosphosphorylation of the Smad linker region. Thus, low FSS activates Smad2/3, while higher FSS blocks nuclear translocation to induce inward artery remodeling, specifically at low FSS. These results are likely relevant to inward remodeling in atherosclerotic vessels, in which Smad2/3 is activated through TGF-ß signaling.


Asunto(s)
Arterias Carótidas/fisiología , Enfermedades de las Arterias Carótidas/prevención & control , Células Endoteliales/fisiología , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Estrés Mecánico , Remodelación Vascular , Animales , Arterias Carótidas/citología , Enfermedades de las Arterias Carótidas/metabolismo , Enfermedades de las Arterias Carótidas/patología , Células Endoteliales/citología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación , Transducción de Señal , Proteína Smad2/genética , Proteína smad3/genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
7.
Mol Genet Genomics ; 298(4): 823-836, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37059908

RESUMEN

Coronavirus 2019 (COVID-19) is a complex disease that affects billions of people worldwide. Currently, effective etiological treatment of COVID-19 is still lacking; COVID-19 also causes damages to various organs that affects therapeutics and mortality of the patients. Surveillance of the treatment responses and organ injury assessment of COVID-19 patients are of high clinical value. In this study, we investigated the characteristic fragmentation patterns and explored the potential in tissue injury assessment of plasma cell-free DNA in COVID-19 patients. Through recruitment of 37 COVID-19 patients, 32 controls and analysis of 208 blood samples upon diagnosis and during treatment, we report gross abnormalities in cfDNA of COVID-19 patients, including elevated GC content, altered molecule size and end motif patterns. More importantly, such cfDNA fragmentation characteristics reflect patient-specific physiological changes during treatment. Further analysis on cfDNA tissue-of-origin tracing reveals frequent tissue injuries in COVID-19 patients, which is supported by clinical diagnoses. Hence, our work demonstrates and extends the translational merit of cfDNA fragmentation pattern as valuable analyte for effective treatment monitoring, as well as tissue injury assessment in COVID-19.


Asunto(s)
COVID-19 , Ácidos Nucleicos Libres de Células , Humanos , COVID-19/diagnóstico , Ácidos Nucleicos Libres de Células/genética
8.
J Virol ; 96(3): e0184221, 2022 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-34817197

RESUMEN

Middle East respiratory syndrome coronavirus (MERS-CoV) is a beta coronavirus that emerged in 2012, causing severe pneumonia and renal failure. MERS-CoV encodes five accessory proteins. Some of them have been shown to interfere with host antiviral immune response. However, the roles of protein 8b in innate immunity and viral virulence was rarely studied. Here, we introduced individual MERS-CoV accessory protein genes into the genome of an attenuated murine coronavirus (Mouse hepatitis virus, MHV), respectively, and found accessory protein 8b could enhance viral replication in vivo and in vitro and increase the lethality of infected mice. RNA-seq analysis revealed that protein 8b could significantly inhibit type I interferon production (IFN-I) and innate immune response in mice infected with MHV expressing protein 8b. We also found that MERS-CoV protein 8b could initiate from multiple internal methionine sites and at least three protein variants were identified. Residues 1-23 of protein 8b was demonstrated to be responsible for increased virulence in vivo. In addition, the inhibitory effect on IFN-I of protein 8b might not contribute to its virulence enhancement as aa1-23 deletion did not affect IFN-I production in vitro and in vivo. Next, we also found that protein 8b was localized to the endoplasmic reticulum (ER)/Golgi membrane in infected cells, which was disrupted by C-terminal region aa 88-112 deletion. This study will provide new insight into the pathogenesis of MERS-CoV infection. IMPORTANCE Multiple coronaviruses (CoV) cause severe respiratory infections and become global public health threats such as SARS-CoV, MERS-CoV, and SARS-CoV-2. Each coronavirus contains different numbers of accessory proteins which show high variability among different CoVs. Accessory proteins are demonstrated to play essential roles in pathogenesis of CoVs. MERS-CoV contains 5 accessory proteins (protein 3, 4a, 4b, 5, 8b), and deletion of all four accessory proteins (protein 3, 4a, 4b, 5), significantly affects MERS-CoV replication and pathogenesis. However, whether ORF8b also regulates MERS-CoV infection is unknown. Here, we constructed mouse hepatitis virus (MHV) recombinant virus expressing MERS-CoV protein 8b and demonstrated protein 8b could significantly enhance the virulence of MHV, which is mediated by N-terminal domain of protein 8b. This study will shed light on the understanding of pathogenesis of MERS-CoV infection.


Asunto(s)
Coronavirus del Síndrome Respiratorio de Oriente Medio/fisiología , Virus de la Hepatitis Murina/fisiología , Dominios y Motivos de Interacción de Proteínas , Proteínas Reguladoras y Accesorias Virales/genética , Animales , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Ratones , Mortalidad , Proteínas Reguladoras y Accesorias Virales/química , Tropismo Viral , Virulencia/genética , Factores de Virulencia/genética
9.
Asia Pac J Clin Nutr ; 32(1): 33-39, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36997483

RESUMEN

BACKGROUND AND OBJECTIVES: Observational studies have shown that energy restriction could be beneficial for controlling bodyweight in patients with polycystic ovary syndrome (PCOS). We aim to compare the effects of a high-protein diet (HPD), a high-protein and high-dietary fiber diet (HPHFD), and a calorie-restricted diet (CRD) on metabolic health and gut microbiota in overweight/obese PCOS patients. METHODS AND STUDY DESIGN: We will enroll a total of 90 overweight/obese PCOS patients into this eight-week open-label randomised controlled trial. Participants will be randomly assigned to three groups: CRD group (energy coefficient 20 kcal/kg.day, water ≥1500 mL, 0.8-1.2 g/kg protein, carbohydrate energize 55-60%, and fat energize 25-30%), HDP group (energy coefficient 20 kcal/kg.day, water ≥1500 mL, and 1.5-2.0 g/kg protein) and HPHFD group (based on the high protein diet with 15 g more dietary fiber supplement). The primary outcome is body weight, body fat percentage, and lean body mass. The secondary outcomes will include changes in blood lipids, inflammation, glucose tolerance, blood pressure, and gut microbiota compositions. Between-group differences in adiposity measurements at baseline will be compared using one-way analysis of variance (ANOVA) or Kruskal-Wallis test when appropriate. Within-group difference after 8-week intervention will be compared using paired t-test or Wilcoxon signed rank test. Between-group differences in adiposity measurements after 8-week diet intervention will be compared using linear mixed model and ANCOVA. The gut microbiota will be analyzed using 16S amplicon sequencing and the sequencing data will be analyzed using the standardized QIIME2 piperline.


Asunto(s)
Microbioma Gastrointestinal , Resistencia a la Insulina , Síndrome del Ovario Poliquístico , Femenino , Humanos , Sobrepeso/complicaciones , Sobrepeso/terapia , Síndrome del Ovario Poliquístico/complicaciones , Síndrome del Ovario Poliquístico/terapia , Pérdida de Peso , Obesidad/complicaciones , Obesidad/terapia , Peso Corporal , Fibras de la Dieta , Ensayos Clínicos Controlados Aleatorios como Asunto
10.
Mol Psychiatry ; 26(10): 6074-6082, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33828237

RESUMEN

Amyloid-ß (Aß) accumulation in the brain is a pivotal event in the pathogenesis of Alzheimer's disease (AD), and its clearance from the brain is impaired in sporadic AD. Previous studies suggest that approximately half of the Aß produced in the brain is cleared by transport into the periphery. However, the mechanism and pathophysiological significance of peripheral Aß clearance remain largely unknown. The kidney is thought to be responsible for Aß clearance, but direct evidence is lacking. In this study, we investigated the impact of unilateral nephrectomy on the dynamic changes in Aß in the blood and brain in both humans and animals and on behavioural deficits and AD pathologies in animals. Furthermore, the therapeutic effects of the diuretic furosemide on Aß clearance via the kidney were assessed. We detected Aß in the kidneys and urine of both humans and animals and found that the Aß level in the blood of the renal artery was higher than that in the blood of the renal vein. Unilateral nephrectomy increased brain Aß deposition; aggravated AD pathologies, including Tau hyperphosphorylation, glial activation, neuroinflammation, and neuronal loss; and aggravated cognitive deficits in APP/PS1 mice. In addition, chronic furosemide treatment reduced blood and brain Aß levels and attenuated AD pathologies and cognitive deficits in APP/PS1 mice. Our findings demonstrate that the kidney physiologically clears Aß from the blood, suggesting that facilitation of Aß clearance via the kidney represents a novel potential therapeutic approach for AD.


Asunto(s)
Enfermedad de Alzheimer , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Riñón/metabolismo , Ratones , Ratones Transgénicos , Presenilina-1/metabolismo
11.
J Org Chem ; 87(5): 2730-2739, 2022 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-35133834

RESUMEN

A catalyst-induced defluorinative, alkylation or metal-free hydroalkylation of gem-difluoroalkenes enabled by visible light was developed. This protocol provided a mild and practical approach to important and novel monofluoroalkenes and difluoromethylene-containing compounds with moderate to excellent yields.

12.
Acta Neuropathol ; 136(4): 525-536, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30074071

RESUMEN

Accumulation of pathological tau is the hallmark of Alzheimer's disease and other tauopathies and is closely correlated with cognitive decline. Clearance of pathological tau from the brain is a major therapeutic strategy for tauopathies. The physiological capacity of the periphery to clear brain-derived tau and its therapeutic potential remain largely unknown. Here, we found that cisterna magna injected 131I-labelled synthetic tau dynamically effluxed from the brain and was mainly cleared from the kidney, blood, and liver in mice; we also found that plasma tau levels in inferior vena cava were lower than those in femoral artery in humans. These findings suggest that tau proteins can efflux out of the brain and be cleared in the periphery under physiological conditions. Next, we showed that lowering blood tau levels via peritoneal dialysis could reduce interstitial fluid (ISF) tau levels in the brain, and tau levels in the blood and ISF were dynamically correlated; furthermore, tau efflux from the brain was accelerated after the addition of another set of peripheral system in a parabiosis model. Finally, we established parabiosis mouse models using tau transgenic mice and their wild-type littermates and found that brain tau levels and related pathologies in parabiotic transgenic mice were significantly reduced after parabiosis, suggesting that chronic enhancement of peripheral tau clearance alleviates pathological tau accumulation and neurodegeneration in the brain. Our study provides the first evidence of physiological clearance of brain-derived pathological tau in the periphery, suggesting that enhancing peripheral tau clearance is a potential therapeutic strategy for tauopathies.


Asunto(s)
Sistema Nervioso Periférico/metabolismo , Tauopatías/metabolismo , Tauopatías/terapia , Proteínas tau/metabolismo , Adulto , Anciano , Animales , Química Encefálica , Cisterna Magna/metabolismo , Líquido Extracelular/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Parabiosis , Diálisis Peritoneal , Distribución Tisular , Vena Cava Inferior/metabolismo , Proteínas tau/genética
13.
J Nucl Cardiol ; 25(6): 2096-2111, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-28695406

RESUMEN

BACKGROUND: Currently, there is no established non-invasive imaging approach to directly evaluate myocardial microcirculatory function in order to diagnose microvascular disease independent of co-existing epicardial disease. In this work, we developed a methodological framework for quantification of intramyocardial blood volume (IMBV) as a novel index of microcirculatory function with SPECT/CT imaging of 99mTc-labeled red blood cells (RBCs). METHODS: Dual-gated myocardial SPECT/CT equilibrium imaging of 99mTc-RBCs was performed on twelve canines under resting conditions. Five correction schemes were studied: cardiac gating with no other corrections (CG), CG with attenuation correction (CG + AC), CG + AC with scatter correction (CG + AC + SC), dual cardiorespiratory gating with AC + SC (DG + AC + SC), and DG + AC + SC with partial volume correction (DG + AC + SC + PVC). Quantification of IMBV using each approach was evaluated in comparison to those obtained from all corrections. The in vivo SPECT estimates of IMBV values were validated against those obtained from ex vivo microCT imaging of the casted hearts. RESULTS: The estimated IMBV with all corrections was 0.15 ± 0.03 for the end-diastolic phase and 0.11 ± 0.03 for the end-systolic phase. The cycle-dependent change in IMBV (ΔIMBV) with all corrections was 23.9 ± 8.6%. Schemes that applied no correction or partial correction resulted in significant over-estimation of IMBV and significant under-underestimation of ΔIMBV. Estimates of IMBV and ΔIMBV using all corrections were consistent with values reported in the literature using invasive techniques. In vivo SPECT estimates of IMBV strongly correlated (R2 ≥ 0.70) with ex vivo measures for the various correction schemes, while the fully corrected scheme yielded the smallest bias. CONCLUSIONS: Non-invasive quantification of IMBV is feasible using 99mTc-RBCs SPECT/CT imaging, however, requires full compensation of physical degradation factors.


Asunto(s)
Volumen Sanguíneo , Circulación Coronaria/fisiología , Microcirculación/fisiología , Tomografía Computarizada por Tomografía Computarizada de Emisión de Fotón Único/métodos , Animales , Perros , Eritrocitos , Femenino , Hemodinámica , Tecnecio , Microtomografía por Rayos X
14.
Proc Natl Acad Sci U S A ; 112(41): 12812-7, 2015 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-26417068

RESUMEN

The contribution of endothelial-derived miR-17∼92 to ischemia-induced arteriogenesis has not been investigated in an in vivo model. In the present study, we demonstrate a critical role for the endothelial-derived miR-17∼92 cluster in shaping physiological and ischemia-triggered arteriogenesis. Endothelial-specific deletion of miR-17∼92 results in an increase in collateral density limbs and hearts and in ischemic limbs compared with control mice, and consequently improves blood flow recovery. Individual cluster components positively or negatively regulate endothelial cell (EC) functions in vitro, and, remarkably, ECs lacking the cluster spontaneously form cords in a manner rescued by miR-17a, -18a, and -19a. Using both in vitro and in vivo analyses, we identified FZD4 and LRP6 as targets of miR-19a/b. Both of these targets were up-regulated in 17∼92 KO ECs compared with control ECs, and both were shown to be targeted by miR-19 using luciferase assays. We demonstrate that miR-19a negatively regulates FZD4, its coreceptor LRP6, and WNT signaling, and that antagonism of miR-19a/b in aged mice improves blood flow recovery after ischemia and reduces repression of these targets. Collectively, these data provide insights into miRNA regulation of arterialization and highlight the importance of vascular WNT signaling in maintaining arterial blood flow.


Asunto(s)
Receptores Frizzled/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , MicroARNs/metabolismo , Familia de Multigenes/fisiología , Neovascularización Fisiológica/fisiología , Vía de Señalización Wnt/fisiología , Animales , Receptores Frizzled/genética , Isquemia/genética , Isquemia/metabolismo , Isquemia/patología , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Ratones , Ratones Noqueados , MicroARNs/genética
15.
Nature ; 479(7371): 122-6, 2011 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-21983962

RESUMEN

PHD2 serves as an oxygen sensor that rescues blood supply by regulating vessel formation and shape in case of oxygen shortage. However, it is unknown whether PHD2 can influence arteriogenesis. Here we studied the role of PHD2 in collateral artery growth by using hindlimb ischaemia as a model, a process that compensates for the lack of blood flow in case of major arterial occlusion. We show that Phd2 (also known as Egln1) haplodeficient (Phd2(+/-)) mice displayed preformed collateral arteries that preserved limb perfusion and prevented tissue necrosis in ischaemia. Improved arteriogenesis in Phd2(+/-) mice was due to an expansion of tissue-resident, M2-like macrophages and their increased release of arteriogenic factors, leading to enhanced smooth muscle cell (SMC) recruitment and growth. Both chronic and acute deletion of one Phd2 allele in macrophages was sufficient to skew their polarization towards a pro-arteriogenic phenotype. Mechanistically, collateral vessel preconditioning relied on the activation of canonical NF-κB pathway in Phd2(+/-) macrophages. These results unravel how PHD2 regulates arteriogenesis and artery homeostasis by controlling a specific differentiation state in macrophages and suggest new treatment options for ischaemic disorders.


Asunto(s)
Arterias/crecimiento & desarrollo , Extremidades/irrigación sanguínea , Isquemia/prevención & control , Macrófagos/metabolismo , Procolágeno-Prolina Dioxigenasa/deficiencia , Procolágeno-Prolina Dioxigenasa/metabolismo , Alelos , Animales , Modelos Animales de Enfermedad , Extremidades/patología , Femenino , Heterocigoto , Homeostasis , Prolina Dioxigenasas del Factor Inducible por Hipoxia , Isquemia/patología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Miocitos del Músculo Liso/citología , FN-kappa B/metabolismo , Necrosis , Fenotipo , Procolágeno-Prolina Dioxigenasa/genética
16.
Proc Natl Acad Sci U S A ; 111(51): 18309-14, 2014 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-25422430

RESUMEN

Posttranscriptional RNA regulation is important in determining the plasticity of cellular phenotypes. However, mechanisms of how RNA binding proteins (RBPs) influence cellular behavior are poorly understood. We show here that the RBP embryonic lethal abnormal vision like 1 (ELAVL1, also know as HuR) regulates the alternative splicing of eukaryotic translation initiation factor 4E nuclear import factor 1 (Eif4enif1), which encodes an eukaryotic translation initiation factor 4E transporter (4E-T) protein and suppresses the expression of capped mRNAs. In the absence of ELAVL1, skipping of exon 11 of Eif4enif1 forms the stable, short isoform, 4E-Ts. This alternative splicing event results in the formation of RNA processing bodies (PBs), enhanced turnover of angiogenic mRNAs, and suppressed sprouting behavior of vascular endothelial cells. Further, endothelial-specific Elavl1 knockout mice exhibited reduced revascularization after hind limb ischemia and tumor angiogenesis in oncogene-induced mammary cancer, resulting in attenuated blood flow and tumor growth, respectively. ELAVL1-regulated alternative splicing of Eif4enif1 leading to enhanced formation of PB and mRNA turnover constitutes a novel posttranscriptional mechanism critical for pathological angiogenesis.


Asunto(s)
Empalme Alternativo/fisiología , Proteínas ELAV/fisiología , Neovascularización Fisiológica/fisiología , Animales , Proteína 1 Similar a ELAV , Exones , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/metabolismo
17.
Proc Natl Acad Sci U S A ; 111(15): 5514-9, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24706887

RESUMEN

Vascular endothelial growth factors (VEGFs) signal via their cognate receptor tyrosine kinases designated VEGFR1-3. We report that the docking protein fibroblast growth factor receptor substrate 2 (FRS2α) plays a critical role in cell signaling via these receptors. In vitro FRS2α regulates VEGF-A and VEGF-C-dependent activation of extracellular signal-regulated receptor kinase signaling and blood and lymphatic endothelial cells migration and proliferation. In vivo endothelial-specific deletion of FRS2α results in the profound impairment of postnatal vascular development and adult angiogenesis, lymphangiogenesis, and arteriogenesis. We conclude that FRS2α is a previously unidentified component of VEGF receptors signaling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de la Membrana/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/fisiología , Transducción de Señal/fisiología , Animales , Movimiento Celular/fisiología , Cartilla de ADN/genética , Células Endoteliales , Perfilación de la Expresión Génica , Vectores Genéticos , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Immunoblotting , Inmunohistoquímica , Inmunoprecipitación , Flujometría por Láser-Doppler , Lentivirus , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Microtomografía por Rayos X
18.
Development ; 140(8): 1720-9, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23533173

RESUMEN

Arteriogenesis requires growth of pre-existing arteriolar collateral networks and determines clinical outcome in arterial occlusive diseases. Factors responsible for the development of arteriolar collateral networks are poorly understood. The Notch ligand Delta-like 4 (Dll4) promotes arterial differentiation and restricts vessel branching. We hypothesized that Dll4 may act as a genetic determinant of collateral arterial networks and functional recovery in stroke and hind limb ischemia models in mice. Genetic loss- and gain-of-function approaches in mice showed that Dll4-Notch signaling restricts pial collateral artery formation by modulating arterial branching morphogenesis during embryogenesis. Adult Dll4(+/-) mice showed increased pial collateral numbers, but stroke volume upon middle cerebral artery occlusion was not reduced compared with wild-type littermates. Likewise, Dll4(+/-) mice showed reduced blood flow conductance after femoral artery occlusion, and, despite markedly increased angiogenesis, tissue ischemia was more severe. In peripheral arteries, loss of Dll4 adversely affected excitation-contraction coupling in arterial smooth muscle in response to vasopressor agents and arterial vessel wall adaption in response to increases in blood flow, collectively contributing to reduced flow reserve. We conclude that Dll4-Notch signaling modulates native collateral formation by acting on vascular branching morphogenesis during embryogenesis. Dll4 furthermore affects tissue perfusion by acting on arterial function and structure. Loss of Dll4 stimulates collateral formation and angiogenesis, but in the context of ischemic diseases such beneficial effects are overruled by adverse functional changes, demonstrating that ischemic recovery is not solely determined by collateral number but rather by vessel functionality.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Isquemia/fisiopatología , Proteínas de la Membrana/metabolismo , Microvasos/embriología , Morfogénesis/fisiología , Neovascularización Fisiológica/fisiología , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Proteínas Adaptadoras Transductoras de Señales , Análisis de Varianza , Animales , Proteínas de Unión al Calcio , Inmunohistoquímica , Isquemia/metabolismo , Ratones , Microvasos/fisiología , Reacción en Cadena en Tiempo Real de la Polimerasa , Flujo Sanguíneo Regional/fisiología , Microtomografía por Rayos X
19.
Microvasc Res ; 106: 57-66, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27009591

RESUMEN

Properly regulated angiogenesis and arteriogenesis are essential for effective wound healing. Tissue injury induces robust new vessel formation and subsequent vessel maturation, which involves vessel regression and remodeling. Although formation of functional vasculature is essential for healing, alterations in vascular structure over the time course of skin wound healing are not well understood. Here, using high-resolution ex vivo X-ray micro-computed tomography (microCT), we describe the vascular network during healing of skin excisional wounds with highly detailed three-dimensional (3D) reconstructed images and associated quantitative analysis. We found that relative vessel volume, surface area and branching number are significantly decreased in wounds from day 7 to days 14 and 21. Segmentation and skeletonization analysis of selected branches from high-resolution images as small as 2.5µm voxel size show that branching orders are decreased in the wound vessels during healing. In histological analysis, we found that the contrast agent fills mainly arterioles, but not small capillaries nor large veins. In summary, high-resolution microCT revealed dynamic alterations of vessel structures during wound healing. This technique may be useful as a key tool in the study of the formation and regression of wound vessels.


Asunto(s)
Angiografía por Tomografía Computarizada/métodos , Neovascularización Fisiológica , Piel/irrigación sanguínea , Piel/diagnóstico por imagen , Cicatrización de Heridas , Heridas y Lesiones/diagnóstico por imagen , Microtomografía por Rayos X , Animales , Arteriolas/diagnóstico por imagen , Arteriolas/fisiopatología , Modelos Animales de Enfermedad , Imagenología Tridimensional , Masculino , Ratones Endogámicos C57BL , Valor Predictivo de las Pruebas , Interpretación de Imagen Radiográfica Asistida por Computador , Factores de Tiempo , Heridas y Lesiones/fisiopatología
20.
Mol Cell ; 32(1): 140-9, 2008 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-18851840

RESUMEN

Mammalian target of rapamycin (mTOR) activity is regulated by assembly of two functionally distinct complexes, mTORC1 and mTORC2. In syndecan-4 (S4) null endothelial cells, mTORC2 activity is reduced, resulting in decreased Akt activation, while mTORC1 activity is increased. Levels of rictor, mLST8, and mSin-1 are unchanged in total cell lysates but decreased in the rafts of S4(-/-) endothelial cells, as is the level of PKCalpha. Expression of myristoylated-PKCalpha in S4(-/-) cells restores rictor, mLST8, and mSin-1 presence in the rafts and rescues Akt phosphorylation. PKCalpha knockdown mimics the effect of S4 deletion on mTORC2 localization and Akt activation. Reduced mTORC2 activity in S4(-/-) endothelial cells results in decreased FoxO1/3a and eNOS phosphorylation, decreased endothelial cell size, and increased arterial blood pressure in S4(-/-) mice. Thus, S4-dependent targeting of PKCalpha to the plasma membrane is required for recruitment of mTORC2 components to the rafts and Akt activation.


Asunto(s)
Proteína Quinasa C-alfa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sindecano-4/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Transporte Biológico Activo , Línea Celular , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Activación Enzimática/efectos de los fármacos , Sustancias de Crecimiento/farmacología , Microdominios de Membrana/metabolismo , Ratones , Ratones Noqueados , Modelos Biológicos , Sindecano-4/deficiencia , Sindecano-4/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA