Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 807
Filtrar
1.
Adv Healthc Mater ; 11(2): e2101873, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34710291

RESUMEN

The principle challenge for engineering viable, cell-laden hydrogel constructs of clinically-relevant size, is rapid vascularization, in order to moderate the finite capacity of passive nutrient diffusion. A multiscale vascular approach, with large open channels and bulk microcapillaries may be an admissible approach to accelerate this process, promoting overall pre-vascularization for long-term viability of constructs. However, the limited availability of bioinks that possess suitable characteristics that support both fabrication of complex architectures and formation of microcapillaries, remains a barrier to advancement in this space. In this study, gelatin-norbornene (Gel-NOR) is investigated as a vascular bioink with tailorable physico-mechanical properties, which promoted the self-assembly of human stromal and endothelial cells into microcapillaries, as well as being compatible with extrusion and lithography-based biofabrication modalities. Gel-NOR constructs containing self-assembled microcapillaries are successfully biofabricated with varying physical architecture (fiber diameter, spacing, and orientation). Both channel sizes and cell types affect the overall structural changes of the printed constructs, where cross-signaling between both human stromal and endothelial cells may be responsible for the reduction in open channel lumen observed over time. Overall, this work highlights an exciting three-way interplay between bioink formulation, construct design, and cell-mediated response that can be exploited towards engineering vascular tissues.


Asunto(s)
Bioimpresión , Capilares , Gelatina , Ingeniería de Tejidos , Capilares/crecimiento & desarrollo , Células Endoteliales , Gelatina/química , Humanos , Hidrogeles/química , Norbornanos/química , Impresión Tridimensional , Andamios del Tejido/química
2.
Dev Dyn ; 250(12): 1704-1716, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34101289

RESUMEN

Intussusceptive angiogenesis (IA) is an important physiological form of angiogenesis in which an existing vessel splits in two by the formation of an intraluminal tissue pillar. The presence of these intraluminal pillars form the hallmark of ongoing IA in growing vascular beds. However, their visualization is technically challenging. The goal of this systematic review was to investigate which techniques are being used to identify intraluminal pillars and to formulate important points to keep in mind when studying IA. A systematic literature search resulted in 154 evaluated articles of which the majority (65%) provided sufficient data to unambiguously demonstrate the presence of intraluminal pillars. Scanning electron microscopy imaging of vascular corrosion casts and serial sectioning of ultrathin sections are the most used techniques. New methods such as serial block face scanning electron microscopy and micro computed tomography (µCT) are gaining importance. Moreover, our results indicate that IA was studied in a variety of animals and tissues. IA is a biologically very relevant form of angiogenesis. Techniques to visualize intraluminal pillars need to have a minimal resolution of 1 µm and should provide information on the 3D-nature of the pillars. Optimally, several techniques are combined to demonstrate ongoing IA.


Asunto(s)
Capilares/crecimiento & desarrollo , Técnicas Citológicas , Neovascularización Fisiológica/fisiología , Animales , Capilares/citología , Capilares/embriología , Técnicas Citológicas/métodos , Técnicas Citológicas/tendencias , Morfogénesis/fisiología
3.
Sci Rep ; 11(1): 11603, 2021 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-34079011

RESUMEN

Blood vessels in the adult mammal exist in a highly organized and stable state. In the ischemic heart, limited expansion capacity of the myocardial vascular bed cannot satisfy demands for oxygen supply and the myocardium eventually undergoes irreversible damage. The predominant contribution of endogenous c-Kit+ cells is understood to be in the development and homeostasis of cardiac endothelial cells, which suggests potential for their targeting in treatments for cardiac ischemic injury. Quiescent cells in other tissues are known to contribute to the long-term maintenance of a cell pool, preserve proliferation capacity and, upon activation, facilitate tissue homeostasis and regeneration in response to tissue injury. Here, we present evidence of a Setd4-expressing quiescent c-Kit+ cell population in the adult mouse heart originating from embryonic stages. Conditional knock-out of Setd4 in c-Kit-CreERT2;Setd4f/f;Rosa26TdTomato mice induced an increase in vascular endothelial cells of capillaries in both neonatal and adult mice. We show that Setd4 regulates quiescence of c-Kit+ cells by the PI3K-Akt-mTOR signaling pathway via H4K20me3 catalysis. In myocardial infarction injured mice, Setd4 knock-out resulted in attenuated cardiomyocyte apoptosis, decreased infarction size and improved cardiac function. Lineage tracing in Setd4-Cre;Rosa26mT/mG mice showed that Setd4+ cells contribute to each cardiac lineage. Overall, Setd4 epigenetically controls c-Kit+ cell quiescence in the adult heart by facilitating heterochromatin formation via H4K20me3. Beyond activation, endogenous quiescent c-Kit+ cells were able to improve cardiac function in myocardial infarction injured mice via the neovascularization of capillaries.


Asunto(s)
Células Endoteliales/metabolismo , Epigénesis Genética , Metiltransferasas/genética , Infarto del Miocardio/genética , Miocitos Cardíacos/metabolismo , Proteínas Proto-Oncogénicas c-kit/genética , Animales , Apoptosis , Capilares/crecimiento & desarrollo , División Celular , Proliferación Celular , Modelos Animales de Enfermedad , Ecocardiografía , Células Endoteliales/citología , Femenino , Histonas/genética , Histonas/metabolismo , Integrasas/genética , Integrasas/metabolismo , Metiltransferasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/citología , Neovascularización Fisiológica , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
4.
Proc Natl Acad Sci U S A ; 118(26)2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34172585

RESUMEN

Capillary networks are essential for distribution of blood flow through the brain, and numerous other homeostatic functions, including neurovascular signal conduction and blood-brain barrier integrity. Accordingly, the impairment of capillary architecture and function lies at the root of many brain diseases. Visualizing how brain capillary networks develop in vivo can reveal innate programs for cerebrovascular growth and repair. Here, we use longitudinal two-photon imaging through noninvasive thinned skull windows to study a burst of angiogenic activity during cerebrovascular development in mouse neonates. We find that angiogenesis leading to the formation of capillary networks originated exclusively from cortical ascending venules. Two angiogenic sprouting activities were observed: 1) early, long-range sprouts that directly connected venules to upstream arteriolar input, establishing the backbone of the capillary bed, and 2) short-range sprouts that contributed to expansion of anastomotic connectivity within the capillary bed. All nascent sprouts were prefabricated with an intact endothelial lumen and pericyte coverage, ensuring their immediate perfusion and stability upon connection to their target vessels. The bulk of this capillary expansion spanned only 2 to 3 d and contributed to an increase of blood flow during a critical period in cortical development.


Asunto(s)
Encéfalo/irrigación sanguínea , Encéfalo/diagnóstico por imagen , Capilares/diagnóstico por imagen , Neuroimagen , Animales , Animales Recién Nacidos , Arteriolas/diagnóstico por imagen , Encéfalo/citología , Capilares/crecimiento & desarrollo , Células Endoteliales/citología , Proteínas Fluorescentes Verdes/metabolismo , Ratones Transgénicos , Neovascularización Fisiológica , Pericitos/citología , Flujo Sanguíneo Regional/fisiología , Factores de Tiempo
5.
Sci Rep ; 11(1): 9644, 2021 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-33958649

RESUMEN

Several studies have suggested that extracellular matrix (ECM) remodeling and the microenvironment are tightly associated with adipogenesis and adipose angiogenesis. In the present study, we demonstrated that transforming growth factor-beta induced (TGFBI) suppresses angiogenesis stimulated by adipocyte-conditioned medium (Ad-CM), both in vitro and in vivo. TGFBI knockout (KO) mice exhibited increased numbers of blood vessels in adipose tissue, and blood vessels from these mice showed enhanced infiltration into Matrigel containing Ad-CM. The treatment of Ad-CM-stimulated SVEC-10 endothelial cells with TGFBI protein reduced migration and tube-forming activity. TGFBI protein suppressed the activation of the Src and extracellular signaling-related kinase signaling pathways of these SVEC-10 endothelial cells. Our findings indicated that TGFBI inhibited adipose angiogenesis by suppressing the activation of Src and ERK signaling pathways, possibly because of the stimulation of the angiogenic activity of endothelial cells.


Asunto(s)
Tejido Adiposo/irrigación sanguínea , Endotelio Vascular/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Neovascularización Fisiológica , Factor de Crecimiento Transformador beta/metabolismo , Tejido Adiposo/metabolismo , Animales , Capilares/crecimiento & desarrollo , Capilares/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
6.
Am J Respir Cell Mol Biol ; 65(1): 81-91, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33784484

RESUMEN

Bronchopulmonary dysplasia (BPD), the most common sequela of preterm birth, is a severe disorder of the lung that is often associated with long-lasting morbidity. A hallmark of BPD is the disruption of alveolarization, whose pathogenesis is incompletely understood. Here, we tested the vascular hypothesis that disordered vascular development precedes the decreased alveolarization associated with BPD. Neonatal mouse pups were exposed to 7, 14, or 21 days of normoxia (21% O2) or hyperoxia (85% O2) with n = 8-11 for each group. The right lungs were fixed by vascular perfusion and investigated by design-based stereology or three-dimensional reconstruction of data sets obtained by serial block-face scanning EM. The alveolar capillary network of hyperoxia-exposed mice was characterized by rarefaction, partially altered geometry, and widening of capillary segments as shown by three-dimensional reconstruction. Stereology revealed that the development of alveolar epithelium and capillary endothelium was decreased in hyperoxia-exposed mice; however, the time course of these effects was different. That the surface area of the alveolar epithelium was smaller in hyperoxia-exposed mice first became evident at Day 14. In contrast, the surface area of the endothelium was reduced in hyperoxia-exposed mouse pups at Day 7. The thickness of the air-blood barrier decreased during postnatal development in normoxic mice, whereas it increased in hyperoxic mice. The endothelium and the septal connective tissue made appreciable contributions to the thickened septa. In conclusion, the present study provides clear support for the idea that the stunted alveolarization follows the disordered microvascular development, thus supporting the vascular hypothesis of BPD.


Asunto(s)
Displasia Broncopulmonar/metabolismo , Capilares/crecimiento & desarrollo , Alveolos Pulmonares/irrigación sanguínea , Alveolos Pulmonares/crecimiento & desarrollo , Animales , Animales Recién Nacidos , Displasia Broncopulmonar/patología , Capilares/patología , Modelos Animales de Enfermedad , Ratones , Alveolos Pulmonares/patología
7.
Invest Ophthalmol Vis Sci ; 62(2): 21, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33595613

RESUMEN

Purpose: Pathological blood vessel growth in the eye is implicated in several diseases that result in vision loss, including age-related macular degeneration and diabetic retinopathy. The limits of current disease therapies have created the need to identify and characterize new antiangiogenic drugs. Here, we identify the secreted chemorepellent semaphorin-3fa (Sema3fa) as an endogenous anti-angiogenic in the eye. Methods: We generated a CRISPR/Cas9 sema3fa zebrafish mutant line, sema3faca304/304. We assessed the retinal and choroidal vasculature in both larval and adult wild-type and sema3fa mutant zebrafish. Results: We find sema3fa mRNA is expressed by the ciliary marginal zone, neural retina, and retinal pigment epithelium of zebrafish larvae as choroidal vascularization emerges and the hyaloid/retinal vasculature is remodeled. The hyaloid vessels of sema3fa mutants develop appropriately but fail to remodel during the larval period, with adult mutants exhibiting a denser network of capillaries in the retinal periphery than seen in wild-type. The choroid vasculature is also defective in that it develops precociously, and aberrant, leaky sprouts are present in the normally avascular outer retina of both sema3faca304/304 larvae and adult fish. Conclusions: Sema3fa is a key endogenous signal for maintaining an avascular retina and preventing pathologic vascularization. Furthermore, we provide a new experimentally accessible model for studying choroid neovascularization (CNV) resulting from primary changes in the retinal environment that lead to downstream vessel infiltration.


Asunto(s)
Capilares/crecimiento & desarrollo , ADN/genética , Degeneración Macular/genética , Proteínas de la Membrana/genética , Mutación , Proteínas del Tejido Nervioso/genética , Epitelio Pigmentado de la Retina/metabolismo , Vasos Retinianos/crecimiento & desarrollo , Animales , Capilares/metabolismo , Coroides/metabolismo , Coroides/patología , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Degeneración Macular/metabolismo , Degeneración Macular/patología , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Epitelio Pigmentado de la Retina/patología , Vasos Retinianos/metabolismo , Pez Cebra
8.
Int J Mol Med ; 47(4)2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33537821

RESUMEN

Endothelial dysfunction and diabetic vascular disease induced by chronic hyperglycemia involve complex interactions among high glucose, long non­coding RNAs (lncRNAs), microRNAs (miRNAs or miRs) and the Ser/Thr kinase AKT. However, the molecular mechanisms underlying the regulatory crosstalk between these have not yet been completely elucidated. Thus, the present study aimed to explore the molecular mechanisms whereby high glucose (HG)­induced lncRNA MIR181A2HG modulates human umbilical vein endothelial cell (HUVEC) proliferation and migration by regulating AKT2 expression. The persistent exposure of HUVECs to HG resulted in MIR181A2HG downregulation and thus reduced its ability to sponge miR­6832­5p, miR­6842­5p and miR­8056, subsequently leading to an increase in miR­6832­5p, miR­6842­5p and miR­8056 levels. Mechanistically, miR­6832­5p, miR­6842­5p and miR­8056 were found to target the 3'UTR of AKT2 mRNA in HUVECs, and the increase in their levels led to a decreased expression of AKT2. Thus, this also led to the suppression of HUVEC proliferation and migration, and the formation of capillary­like structures. Moreover, the suppression of HUVEC proliferation and migration induced by MIR181A2HG downregulation was accompanied by changes in glucose metabolism. On the whole, the present study demonstrates that the downregulation of lncRNA MIR181A2HG by HG impairs HUVEC proliferation and migration by dysregulating the miRNA/AKT2 axis. The MIR181A2HG/miRNA/AKT2 regulatory axis may thus be a potential therapeutic target for HG­induced endothelial dysfunction.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Regulación hacia Abajo/genética , Glucosa/toxicidad , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , MicroARNs/genética , Proteínas Proto-Oncogénicas c-akt/genética , ARN Largo no Codificante/genética , Regiones no Traducidas 3'/genética , Secuencia de Bases , Capilares/efectos de los fármacos , Capilares/crecimiento & desarrollo , Proliferación Celular/efectos de los fármacos , Glucosa/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Modelos Biológicos , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Largo no Codificante/metabolismo
9.
Microcirculation ; 28(4): e12677, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33417723

RESUMEN

OBJECTIVE: Adequacy of the microcirculation is essential for maintaining repetitive skeletal muscle function while avoiding fatigue. It is unclear, however, whether capillary remodelling after different angiogenic stimuli is comparable in terms of vessel distribution and consequent functional adaptations. We determined the physiological consequences of two distinct mechanotransductive stimuli: (1) overload-mediated abluminal stretch (OV); (2) vasodilator-induced shear stress (prazosin, PR). METHODS: In situ EDL fatigue resistance was determined after 7 or 14 days of intervention, in addition to measurements of femoral artery flow. Microvascular composition (muscle histology) and oxidative capacity (citrate synthase activity) were quantified, and muscle PO2 calculated using advanced mathematical modelling. RESULTS: Compared to controls, capillary-to-fiber ratio was higher after OV14 (134%, p < .001) and PR14 (121%, p < .05), although fatigue resistance only improved after overload (7 days: 135%, 14 days: 125%, p < .05). In addition, muscle overload improved local capillary supply indices and reduced CS activity, while prazosin treatment failed to alter either index of aerobic capacity. CONCLUSION: Targeted capillary growth in response to abluminal stretch is a potent driver of improved muscle fatigue resistance, while shear stress-driven angiogenesis has no beneficial effect on muscle function. In terms of capillarity, more is not necessarily better.


Asunto(s)
Antagonistas de Receptores Adrenérgicos alfa 1 , Capilares , Actividad Motora , Músculo Esquelético , Neovascularización Fisiológica , Prazosina , Antagonistas de Receptores Adrenérgicos alfa 1/farmacología , Animales , Fenómenos Biomecánicos/efectos de los fármacos , Fenómenos Biomecánicos/fisiología , Capilares/efectos de los fármacos , Capilares/crecimiento & desarrollo , Capilares/fisiología , Estimulación Eléctrica , Masculino , Microcirculación/efectos de los fármacos , Microcirculación/fisiología , Microvasos/efectos de los fármacos , Microvasos/fisiología , Modelos Animales , Actividad Motora/fisiología , Fatiga Muscular/efectos de los fármacos , Fatiga Muscular/fisiología , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiología , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/fisiología , Consumo de Oxígeno/efectos de los fármacos , Consumo de Oxígeno/fisiología , Prazosina/farmacología , Ratas , Ratas Wistar
10.
Int J Artif Organs ; 44(2): 130-138, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32611278

RESUMEN

Vascularization of tissue engineered implants is crucial for their survival and integration in the recipient's body. Pre-vascularized, fibrin-based implants offer a solution since low concentration fibrin hydrogels (1 mg/mL) have been shown to promote tube formation of endothelial cells in co-culture with adipogenic stem cells. However, higher fibrinogen concentrations (> 20 mg/mL) enabling the fabrication of stable implants are necessary.We here characterized fibrin gels of 1-30 mg/mL for their rheological properties and whether they support tube formation of endothelial cell-adipogenic stem cell co-cultures for up to 7 days. Moreover, 20 mg/mL gels containing preformed channels and endothelial cell-adipogenic stem cell co-culture were perfused continuously in a customized flow chamber with 3.9 dyn/cm2 for 12 days and analyzed for capillary formation.Rheology of fibrin gels showed increasing stability proportional to fibrinogen concentration with 20 mg/mL gels having a storage module of 465 Pa. Complex tube networks stable for 7 days were observed at 1-5 mg/mL gels whereas higher concentrations showed initial sprouting only. However, perfusion of 20 mg/mL fibrin gels resulted in endothelialized pore formation in several layers of the gel with endothelial cell-adipogenic stem cell co-culture.Thus, perfusion supports the formation of capillary-like structures in fibrin gels that are too dense for spontaneous tube formation under static conditions. Future studies are necessary to further increase pore density and to investigate proper nutrition of tissue-specific target cells in the scaffold.


Asunto(s)
Fibrina/farmacología , Regeneración Tisular Dirigida/métodos , Hidrogeles/farmacología , Repitelización/fisiología , Ingeniería de Tejidos , Andamios del Tejido , Implantes Absorbibles , Capilares/crecimiento & desarrollo , Humanos , Perfusión/métodos , Prótesis e Implantes/normas , Reología , Ingeniería de Tejidos/instrumentación , Ingeniería de Tejidos/métodos
11.
Genes (Basel) ; 11(11)2020 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-33137935

RESUMEN

BACKGROUND: We demonstrated that the transduction of Wnt11 into mesenchymal stem cells (MSCs) (MSCWnt11) promotes these cells differentiation into cardiac phenotypes. In the present study, we investigated the paracrine effects of MSCWnt11 on cardiac function and angiogenesis. METHODS AND RESULTS: Conditioned medium was collected from MSCWnt11 (CdMWnt11) and their control cells (CdMGFP). CdMWnt11, especially obtained from MSCWnt11 exposed to hypoxia, significantly promoted human umbilical vein endothelial cells (HUVECs) migration and increased capillary-like tube (CLT) formation, which was blocked by Wnt11 neutralizing antibody. Wnt11 protein was significantly higher in CdMWnt11 compared to that in CdMGFP. Directly treating HUVECs with recombinant Wnt11 protein significantly increased CLT formation, which was abrogated by treating cells with the JNK inhibitor SP600125, as well as the PKC inhibitor Calphostin-C. Moreover, the transfection of Wnt11 to HUVECs (HWnt11) significantly increased CLT formation and HUVEC migration, as well as upregulated p-pan-PKC and p-JNK expression. Injection of CdMWnt11 into the peri-infarct region in a rat acute myocardial infarction (AMI) model significantly improved cardiac function, reduced infarct size, and increased myocardial blood flow and blood vessel density in the ischemic area. CONCLUSION: Wnt11 released from MSCWnt11 increased angiogenesis and improved cardiac function via non-canonical Wnt-PKC-JNK dependent pathways.


Asunto(s)
Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Neovascularización Fisiológica , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , Animales , Capilares/citología , Capilares/crecimiento & desarrollo , Capilares/metabolismo , Células Cultivadas , Medios de Cultivo Condicionados , Modelos Animales de Enfermedad , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Sistema de Señalización de MAP Quinasas , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Neovascularización Fisiológica/genética , Proteína Quinasa C/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Wnt/antagonistas & inhibidores , Proteínas Wnt/genética , Vía de Señalización Wnt/genética
12.
Reprod Domest Anim ; 55(11): 1479-1489, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32762098

RESUMEN

Non-infectious prenatal mortality severely affects the porcine industry, with pathological placentation as a likely key reason. Previous studies have demonstrated that peroxisome proliferator-activated receptor gamma (PPARγ) deficiency causes defects in the uteroplacental vasculature and induces embryonic losses in mice. However, its role in porcine placental angiogenesis remains unclear. In the present study, PPARγ expression was investigated in porcine uteroplacental tissues at gestational day (GD) 25, GD40 and GD70 via quantitative polymerase chain reaction (qPCR), Western blot and immunohistochemistry (IHC). Moreover, the roles of PPARγ in porcine placental angiogenesis were investigated using a cell model of porcine umbilical vein endothelial cells (PUVECs) to conduct proliferation, migration and tube formation assays in vitro and a mouse xenograft model to assess capillary formation in vivo. The results showed that PPARγ was mainly located in the glandular epithelium, trophoblast, amniotic chorion epithelium and vascular endothelium, as indicated by the higher expression levels at GD25 and GD40 than at GD70 in endometrium and by higher expression levels at GD40 and GD70 than at GD25 in placenta. Moreover, PPARγ expression was significantly downregulated in placenta with dead foetus. In PUVECs, knocking out PPARγ significantly inhibited proliferation, migration and tube formation in vitro and inhibited capillary formation in mouse xenografts in vivo by blocking S-phase, promoting apoptosis and downregulating the angiogenic factors of VEGF and its receptors. Overall, the spatiotemporal heterogeneity of PPARγ expression in porcine uteroplacental tissue suggests its vital role in endometrial remodelling and placental angiogenesis, and PPARγ regulates placental angiogenesis through VEGF-mediated signalling.


Asunto(s)
PPAR gamma/metabolismo , Placenta/irrigación sanguínea , Sus scrofa/fisiología , Animales , Capilares/crecimiento & desarrollo , Línea Celular , Femenino , Xenoinjertos , Ratones Endogámicos C57BL , Neovascularización Fisiológica , PPAR gamma/genética , Placenta/metabolismo , Embarazo , Transducción de Señal , Útero/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
13.
J Neurosci ; 40(34): 6503-6521, 2020 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-32661024

RESUMEN

Microglia, a resident CNS macrophage, are dynamic cells, constantly extending and retracting their processes as they contact and functionally regulate neurons and other glial cells. There is far less known about microglia-vascular interactions, particularly under healthy steady-state conditions. Here, we use the male and female mouse cerebral cortex to show that a higher percentage of microglia associate with the vasculature during the first week of postnatal development compared with older ages and that the timing of these associations is dependent on the fractalkine receptor (CX3CR1). Similar developmental microglia-vascular associations were detected in the human brain. Using live imaging in mice, we found that juxtavascular microglia migrated when microglia are actively colonizing the cortex and became stationary by adulthood to occupy the same vascular space for nearly 2 months. Further, juxtavascular microglia at all ages associate with vascular areas void of astrocyte endfeet, and the developmental shift in microglial migratory behavior along vessels corresponded to when astrocyte endfeet more fully ensheath vessels. Together, our data provide a comprehensive assessment of microglia-vascular interactions. They support a mechanism by which microglia use the vasculature to migrate within the developing brain parenchyma. This migration becomes restricted on the arrival of astrocyte endfeet such that juxtavascular microglia become highly stationary and stable in the mature cortex.SIGNIFICANCE STATEMENT We report the first extensive analysis of juxtavascular microglia in the healthy, developing, and adult brain. Live imaging revealed that juxtavascular microglia within the cortex are highly motile and migrate along vessels as they are colonizing cortical regions. Using confocal, expansion, super-resolution, and electron microscopy, we determined that microglia associate with the vasculature at all ages in areas lacking full astrocyte endfoot coverage and motility of juxtavascular microglia ceases as astrocyte endfeet more fully ensheath the vasculature. Our data lay the fundamental groundwork to investigate microglia-astrocyte cross talk and juxtavascular microglial function in the healthy and diseased brain. They further provide a potential mechanism by which vascular interactions facilitate microglial colonization of the brain to later regulate neural circuit development.


Asunto(s)
Corteza Cerebral/irrigación sanguínea , Corteza Cerebral/crecimiento & desarrollo , Microglía/fisiología , Animales , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/metabolismo , Capilares/crecimiento & desarrollo , Capilares/ultraestructura , Corteza Cerebral/ultraestructura , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Microglía/ultraestructura , Corteza Somatosensorial/metabolismo
14.
Int Immunopharmacol ; 86: 106687, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32570033

RESUMEN

OBJECTIVE: KN motif and ankyrin repeat domains 2 (KANK2) may inhibit the activation of (NF-kappaB) p65, which plays a role in myocardial injury. Thus, our study aims to discover the effect of KANK2 on myocardial infarction (MI) induced by ligating the left anterior descending coronary artery (LAD) through regulating NF-κB p65 in vivo. METHODS: MI rats underwent LAD ligation were administered with intramyocardial injections of KANK2/Control activation plasmids. Six weeks after MI, pressure-volume (P/V) loops was used to investigate the cardiac function of rats, then the following detections were performed, including TTC staining, HE staining, immunofluorescence, Masson' s trichrome staining, ELISA assay, TUNEL staining, immunohistochemistry, qRT-PCR and Western blotting. RESULTS: MI rats decreased in maximum pressure (pmax), ejection fraction (EF%), peak rate of pressure rise (dpdtmax) and decline (-dpdtmax) with increased end diastolic pressure (EDP), which was partially reversed by KANK2 overexpression. Besides, KANK2 CRISPR activation plasmids reduced infarct size with less collagen fiber proliferation and neutrophil infiltration in infarct tissues, as well as suppressed pro-inflammatory factors expressions in MI rats. Moreover, injection of KANK2 activation plasmid decreased collagen deposition, aggravated cardiomyocyte apoptosis, enhanced the capillary density, and increased the expressions of VEGF and bFGF in the infarct and peri-infarct regions of MI rats. KANK2 lowered myocardial NF-κB p65 expression in MI rats. CONCLUSION: KANK2 may play its therapeutic role in MI through improving cardiac function, decreasing myocardial collagen deposition, reducing cardiomyocyte apoptosis, and increasing angiogenesis, which might be associated with the reduction of NF-κB p65 expression.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Infarto del Miocardio/metabolismo , Factor de Transcripción ReIA/antagonistas & inhibidores , Animales , Apoptosis/genética , Capilares/crecimiento & desarrollo , Colágeno/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Infarto del Miocardio/patología , Miocitos Cardíacos/metabolismo , Ratas Sprague-Dawley , Factor de Transcripción ReIA/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Función Ventricular Izquierda
15.
Sci Rep ; 10(1): 9291, 2020 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-32518266

RESUMEN

Tissue engineering is an emerging and promising concept to replace or cure failing organs, but its clinical translation currently encounters issues due to the inability to quickly produce inexpensive thick tissues, which are necessary for many applications. To circumvent this problem, we postulate that cells secrete the optimal cocktail required to promote angiogenesis when they are placed in physiological conditions where their oxygen supply is reduced. Thus, dermal fibroblasts were cultivated under hypoxia (2% O2) to condition their cell culture medium. The potential of this conditioned medium was tested for human umbilical vein endothelial cell proliferation and for their ability to form capillary-like networks into fibrin gels. The medium conditioned by dermal fibroblasts under hypoxic conditions (DF-Hx) induced a more significant proliferation of endothelial cells compared to medium conditioned by dermal fibroblasts under normoxic conditions (DF-Nx). In essence, doubling time for endothelial cells in DF-Hx was reduced by 10.4% compared to DF-Nx after 1 week of conditioning, and by 20.3% after 2 weeks. The DF-Hx allowed the formation of more extended and more structured capillary-like networks than DF-Nx or commercially available medium, paving the way to further refinements.


Asunto(s)
Anaerobiosis/fisiología , Capilares/crecimiento & desarrollo , Medios de Cultivo Condicionados/farmacología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Ingeniería de Tejidos/métodos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Fibrina , Fibroblastos/metabolismo , Geles , Humanos , Neovascularización Fisiológica/fisiología , Trasplante de Órganos/métodos , Oxígeno/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
16.
Cells ; 9(4)2020 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-32316253

RESUMEN

Beyond their role in pathogen recognition and the initiation of immune defense, Toll-like receptors (TLRs) are known to be involved in various vascular processes in health and disease. We investigated the potential of the lipopeptide and TLR2/6 ligand macrophage activating protein of 2-kDA (MALP-2) to promote blood flow recovery in mice. Hypercholesterolemic apolipoprotein E (Apoe)-deficient mice were subjected to microsurgical ligation of the femoral artery. MALP-2 significantly improved blood flow recovery at early time points (three and seven days), as assessed by repeated laser speckle imaging, and increased the growth of pre-existing collateral arteries in the upper hind limb, along with intimal endothelial cell proliferation in the collateral wall and pericollateral macrophage accumulation. In addition, MALP-2 increased capillary density in the lower hind limb. MALP-2 enhanced endothelial nitric oxide synthase (eNOS) phosphorylation and nitric oxide (NO) release from endothelial cells and improved the experimental vasorelaxation of mesenteric arteries ex vivo. In vitro, MALP-2 led to the up-regulated expression of major endothelial adhesion molecules as well as their leukocyte integrin receptors and consequently enhanced the endothelial adhesion of leukocytes. Using the experimental approach of femoral artery ligation (FAL), we achieved promising results with MALP-2 to promote peripheral blood flow recovery by collateral artery growth.


Asunto(s)
Circulación Sanguínea/efectos de los fármacos , Arteria Femoral/efectos de los fármacos , Lipopéptidos/farmacología , Macrófagos/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 6/metabolismo , Animales , Apolipoproteínas E/deficiencia , Capilares/efectos de los fármacos , Capilares/crecimiento & desarrollo , Proliferación Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Arteria Femoral/diagnóstico por imagen , Arteria Femoral/patología , Arteria Femoral/cirugía , Inmunohistoquímica , Imágenes de Contraste de Punto Láser , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/metabolismo , Fosforilación , Vasodilatación/efectos de los fármacos
17.
Sci Rep ; 10(1): 6578, 2020 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-32313031

RESUMEN

Skeletal muscle capillarization is a determining factor in gas and metabolite exchange, while its impairments may contribute to the development of sarcopenia. Studies on the potential of resistance training (RT) to induce angiogenesis in older muscles have been inconclusive, and effects of sequential endurance training (ET) and RT on capillarization are unknown. Healthy older men (66.5 ± 3.8 years) were engaged in either 12 weeks of habitual course observation (HC) followed by 12 weeks of RT (n = 8), or 12 weeks of high-intensity interval training (HIIT) followed by 12 weeks of RT (n = 9). At baseline, following 12 and 24 weeks, m. vastus lateralis biopsies were obtained. (Immuno-)histochemistry was used to assess indices of muscle fiber capillarization, muscle fiber morphology and succinate dehydrogenase (SDH) activity. Single periods of RT and HIIT resulted in similar improvements in capillarization and SDH activity. During RT following HIIT, improved capillarization and SDH activity, as well as muscle fiber morphology remained unchanged. The applied RT and HIIT protocols were thus similarly effective in enhancing capillarization and oxidative enzyme activity and RT effectively preserved HIIT-induced adaptations of these parameters. Hence, both, RT and HIIT, are valid training modalities for older men to improve skeletal muscle vascularization.


Asunto(s)
Envejecimiento/fisiología , Ejercicio Físico , Músculo Esquelético/fisiología , Entrenamiento de Fuerza , Adaptación Fisiológica , Anciano , Envejecimiento/genética , Composición Corporal/fisiología , Capilares/crecimiento & desarrollo , Capilares/fisiología , Femenino , Voluntarios Sanos , Humanos , Masculino , Fibras Musculares Esqueléticas/metabolismo , Factores de Riesgo , Sarcopenia/fisiopatología
18.
Sci Rep ; 10(1): 3842, 2020 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-32123209

RESUMEN

The development and physiologic role of small intestine (SI) vasculature is poorly studied. This is partly due to a lack of targetable, organ-specific markers for in vivo studies of two critical tissue components: endothelium and stroma. This challenge is exacerbated by limitations of traditional cell culture techniques, which fail to recapitulate mechanobiologic stimuli known to affect vessel development. Here, we construct and characterize a 3D in vitro microfluidic model that supports the growth of patient-derived intestinal subepithelial myofibroblasts (ISEMFs) and endothelial cells (ECs) into perfused capillary networks. We report how ISEMF and EC-derived vasculature responds to physiologic parameters such as oxygen tension, cell density, growth factors, and pharmacotherapy with an antineoplastic agent (Erlotinib). Finally, we demonstrate effects of ISEMF and EC co-culture on patient-derived human intestinal epithelial cells (HIECs), and incorporate perfused vasculature into a gut-on-a-chip (GOC) model that includes HIECs. Overall, we demonstrate that ISEMFs possess angiogenic properties as evidenced by their ability to reliably, reproducibly, and quantifiably facilitate development of perfused vasculature in a microfluidic system. We furthermore demonstrate the feasibility of including perfused vasculature, including ISEMFs, as critical components of a novel, patient-derived, GOC system with translational relevance as a platform for precision and personalized medicine research.


Asunto(s)
Capilares/crecimiento & desarrollo , Técnicas de Cocultivo/instrumentación , Intestino Delgado/citología , Dispositivos Laboratorio en un Chip , Miofibroblastos/citología , Humanos , Miofibroblastos/metabolismo , Oxígeno/metabolismo , Perfusión
19.
Cell Mol Biol Lett ; 25: 3, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32042281

RESUMEN

BACKGROUND: Precise coordination of cytoskeletal components and dynamic control of cell adhesion and migration are required for crucial cell processes such as differentiation and morphogenesis. We investigated the potential involvement of αII-spectrin, a ubiquitous scaffolding element of the membrane skeleton, in the adhesion and angiogenesis mechanism. METHODS: The cell models were primary human umbilical vein endothelial cells (HUVECs) and a human dermal microvascular endothelial cell line (HMEC-1). After siRNA- and shRNA-mediated knockdown of αII-spectrin, we assessed its expression and that of its partners and adhesion proteins using western blotting. The phenotypes of the control and spectrin-depleted cells were examined using immunofluorescence and video microscopy. Capillary tube formation was assessed using the thick gel Matrigel matrix-based method and a microscope equipped with a thermostatic chamber and a Nikon Biostation System camera. RESULTS: Knockdown of αII-spectrin leads to: modified cell shape; actin cytoskeleton organization with the presence of peripheral actin patches; and decreased formation of stress fibers. Spectrin deficiency affects cell adhesion on laminin and fibronectin and cell motility. This included modification of the localization of adhesion molecules, such as αVß3- and α5-integrins, and organization of adhesion structures, such as focal points. Deficiency of αII-spectrin can also affect the complex mechanism of in vitro capillary tube formation, as demonstrated in a model of angiogenesis. Live imaging revealed that impairment of capillary tube assembly was mainly associated with a significant decrease in cell projection length and stability. αII-spectrin depletion is also associated with significantly decreased expression of three proteins involved in capillary tube formation and assembly: VE-cadherin, MCAM and ß3-integrin. CONCLUSION: Our data confirm the role of αII-spectrin in the control of cell adhesion and spreading. Moreover, our findings further support the participation of αII-spectrin in capillary tube formation in vitro through control of adhesion molecules, such as integrins. This indicates a new function of αII-spectrin in angiogenesis.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Capilares/metabolismo , Adhesión Celular/fisiología , Células Endoteliales/metabolismo , Neovascularización Fisiológica , Espectrina/metabolismo , Antígenos CD/metabolismo , Antígeno CD146/metabolismo , Cadherinas/metabolismo , Capilares/crecimiento & desarrollo , Adhesión Celular/genética , Diferenciación Celular/genética , Movimiento Celular/genética , Forma de la Célula , Células Endoteliales/citología , Fibronectinas/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Cadenas alfa de Integrinas/metabolismo , Integrina beta3/metabolismo , Laminina/metabolismo , Morfogénesis/genética , Morfogénesis/fisiología , Neovascularización Fisiológica/genética , ARN Interferente Pequeño , Espectrina/deficiencia , Espectrina/genética , Fibras de Estrés/metabolismo
20.
J Cereb Blood Flow Metab ; 40(12): 2475-2490, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-31903837

RESUMEN

Vessel loss in the aging brain is commonly reported, yet important questions remain concerning whether there are regional vulnerabilities and what mechanisms could account for these regional differences, if they exist. Here we imaged and quantified vessel length, tortuosity and width in 15 brain regions in young adult and aged mice. Our data indicate that vessel loss was most pronounced in white matter followed by cortical, then subcortical grey matter regions, while some regions (visual cortex, amygdala, thalamus) showed no decline with aging. Regions supplied by the anterior cerebral artery were more vulnerable to loss than those supplied by middle or posterior cerebral arteries. Vessel width and tortuosity generally increased with age but neither reliably predicted regional vessel loss. Since capillaries are naturally prone to plugging and prolonged obstructions often lead to vessel pruning, we hypothesized that regional susceptibilities to plugging could help predict vessel loss. By mapping the distribution of microsphere-induced capillary obstructions, we discovered that regions with a higher density of persistent obstructions were more likely to show vessel loss with aging and vice versa. These findings indicate that age-related vessel loss is region specific and can be explained, at least partially, by regional susceptibilities to capillary plugging.


Asunto(s)
Isquemia Encefálica/patología , Encéfalo/irrigación sanguínea , Capilares/patología , Corteza Cerebral/patología , Demencia Vascular/patología , Rarefacción Microvascular/fisiopatología , Envejecimiento/fisiología , Animales , Encéfalo/patología , Isquemia Encefálica/fisiopatología , Capilares/crecimiento & desarrollo , Capilares/fisiopatología , Corteza Cerebral/irrigación sanguínea , Corteza Cerebral/fisiopatología , Circulación Cerebrovascular/fisiología , Demencia Vascular/etiología , Demencia Vascular/fisiopatología , Femenino , Sustancia Gris/irrigación sanguínea , Sustancia Gris/patología , Sustancia Gris/fisiopatología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Valor Predictivo de las Pruebas , Sustancia Blanca/irrigación sanguínea , Sustancia Blanca/patología , Sustancia Blanca/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...