Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 890
Filtrar
1.
Sci Rep ; 14(1): 13287, 2024 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-38858395

RESUMEN

Clinical outcomes of arteriovenous fistulae (AVF) for hemodialysis remain inadequate since biological mechanisms of AVF maturation and failure are still poorly understood. Aortocaval fistula creation (AVF group) or a sham operation (sham group) was performed in C57BL/6 mice. Venous limbs were collected on postoperative day 7 and total RNA was extracted for high throughput RNA sequencing and bioinformatic analysis. Genes in metabolic pathways were significantly downregulated in the AVF, whereas significant sex differences were not detected. Since gene expression patterns among the AVF group were heterogenous, the AVF group was divided into a 'normal' AVF (nAVF) group and an 'outliers' (OUT) group. The gene expression patterns of the nAVF and OUT groups were consistent with previously published data showing venous adaptive remodeling, whereas enrichment analyses showed significant upregulation of metabolism, inflammation and coagulation in the OUT group compared to the nAVF group, suggesting the heterogeneity during venous remodeling reflects early gene expression changes that may correlate with AVF maturation or failure. Early detection of these processes may be a translational strategy to predict fistula failure and reduce patient morbidity.


Asunto(s)
Derivación Arteriovenosa Quirúrgica , Ratones Endogámicos C57BL , Remodelación Vascular , Animales , Ratones , Masculino , Remodelación Vascular/genética , Femenino , Regulación hacia Abajo/genética , Venas/metabolismo , Diálisis Renal , Fístula Arteriovenosa/genética , Fístula Arteriovenosa/metabolismo , Fístula Arteriovenosa/patología , Regulación de la Expresión Génica , Perfilación de la Expresión Génica
2.
Cells ; 13(10)2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38786017

RESUMEN

Arteries and veins develop different types of occlusive diseases and respond differently to injury. The biological reasons for this discrepancy are not well understood, which is a limiting factor for the development of vein-targeted therapies. This study contrasts human peripheral arteries and veins at the single-cell level, with a focus on cell populations with remodeling potential. Upper arm arteries (brachial) and veins (basilic/cephalic) from 30 organ donors were compared using a combination of bulk and single-cell RNA sequencing, proteomics, flow cytometry, and histology. The cellular atlases of six arteries and veins demonstrated a 7.8× higher proportion of contractile smooth muscle cells (SMCs) in arteries and a trend toward more modulated SMCs. In contrast, veins showed a higher abundance of endothelial cells, pericytes, and macrophages, as well as an increasing trend in fibroblasts. Activated fibroblasts had similar proportions in both types of vessels but with significant differences in gene expression. Modulated SMCs and activated fibroblasts were characterized by the upregulation of MYH10, FN1, COL8A1, and ITGA10. Activated fibroblasts also expressed F2R, POSTN, and COMP and were confirmed by F2R/CD90 flow cytometry. Activated fibroblasts from veins were the top producers of collagens among all fibroblast populations from both types of vessels. Venous fibroblasts were also highly angiogenic, proinflammatory, and hyper-responders to reactive oxygen species. Differences in wall structure further explain the significant contribution of fibroblast populations to remodeling in veins. Fibroblasts are almost exclusively located outside the external elastic lamina in arteries, while widely distributed throughout the venous wall. In line with the above, ECM-targeted proteomics confirmed a higher abundance of fibrillar collagens in veins vs. more basement ECM components in arteries. The distinct cellular compositions and transcriptional programs of reparative populations in arteries and veins may explain differences in acute and chronic wall remodeling between vessels. This information may be relevant for the development of antistenotic therapies.


Asunto(s)
Arterias , Miocitos del Músculo Liso , Análisis de la Célula Individual , Remodelación Vascular , Venas , Humanos , Arterias/metabolismo , Venas/metabolismo , Miocitos del Músculo Liso/metabolismo , Fibroblastos/metabolismo , Masculino , Femenino , Persona de Mediana Edad
3.
Nature ; 627(8002): 165-173, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38326613

RESUMEN

The arachnoid barrier delineates the border between the central nervous system and dura mater. Although the arachnoid barrier creates a partition, communication between the central nervous system and the dura mater is crucial for waste clearance and immune surveillance1,2. How the arachnoid barrier balances separation and communication is poorly understood. Here, using transcriptomic data, we developed transgenic mice to examine specific anatomical structures that function as routes across the arachnoid barrier. Bridging veins create discontinuities where they cross the arachnoid barrier, forming structures that we termed arachnoid cuff exit (ACE) points. The openings that ACE points create allow the exchange of fluids and molecules between the subarachnoid space and the dura, enabling the drainage of cerebrospinal fluid and limited entry of molecules from the dura to the subarachnoid space. In healthy human volunteers, magnetic resonance imaging tracers transit along bridging veins in a similar manner to access the subarachnoid space. Notably, in neuroinflammatory conditions such as experimental autoimmune encephalomyelitis, ACE points also enable cellular trafficking, representing a route for immune cells to directly enter the subarachnoid space from the dura mater. Collectively, our results indicate that ACE points are a critical part of the anatomy of neuroimmune communication in both mice and humans that link the central nervous system with the dura and its immunological diversity and waste clearance systems.


Asunto(s)
Aracnoides , Encéfalo , Duramadre , Animales , Humanos , Ratones , Aracnoides/anatomía & histología , Aracnoides/irrigación sanguínea , Aracnoides/inmunología , Aracnoides/metabolismo , Transporte Biológico , Encéfalo/anatomía & histología , Encéfalo/irrigación sanguínea , Encéfalo/inmunología , Encéfalo/metabolismo , Duramadre/anatomía & histología , Duramadre/irrigación sanguínea , Duramadre/inmunología , Duramadre/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Perfilación de la Expresión Génica , Imagen por Resonancia Magnética , Ratones Transgénicos , Espacio Subaracnoideo/anatomía & histología , Espacio Subaracnoideo/irrigación sanguínea , Espacio Subaracnoideo/inmunología , Espacio Subaracnoideo/metabolismo , Líquido Cefalorraquídeo/metabolismo , Venas/metabolismo
4.
Sci Rep ; 13(1): 22786, 2023 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-38123618

RESUMEN

Newly created arteriovenous fistulas (AVFs) often fail to mature for dialysis use due to disturbed blood flow at and near the AVF anastomosis. The disturbed flow inhibits the endothelial nitric oxide synthase (NOS3) pathway, thus decreasing the production of nitric oxide, a vasodilator. Previously, our group reported that NOS3 expression levels affect AVF lumen size in a mouse model. In this study, we performed MRI-based computational fluid dynamics simulations to investigate the hemodynamical parameters (velocity, wall shear stress (WSS), and vorticity) in a mouse AVF model at day 7 and day 21 post-AVF creation using three NOS3 strains: overexpression (OE), knockout (KO), and wild-type (WT) control. This study is the first to reveal hemodynamics over time in mouse AVFs, consider spatial heterogeneity along the vein, and reveal the effect of NOS3 on the natural history of mouse AVF hemodynamics. From day 7 to day 21, OE has smoother streamlines and had significantly lower vorticity and WSS than WT and KO, suggesting that WSS was attempting to return to pre-surgery baseline, respectively. Our results conclude that the overexpression of NOS3 leads to desired optimal hemodynamics during AVF remodeling. Future studies can investigate enhancing the NOS3 pathway to improve AVF development.


Asunto(s)
Fístula Arteriovenosa , Óxido Nítrico Sintasa de Tipo III , Animales , Ratones , Modelos Animales de Enfermedad , Hemodinámica/fisiología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Diálisis Renal , Venas/metabolismo
5.
Sci Rep ; 13(1): 4483, 2023 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-36934164

RESUMEN

Endothelial dysfunction is an early event of vascular injury defined by a proinflammatory and procoagulant endothelial cell (EC) phenotype. Although endothelial glycocalyx disruption is associated with vascular damage, how various inflammatory stimuli affect the glycocalyx and whether arterial and venous cells respond differently is unknown. Using a 3D round-channel microfluidic system we investigated the endothelial glycocalyx, particularly heparan sulfate (HS), on porcine arterial and venous ECs. Heparan sulfate (HS)/glycocalyx expression was observed already under static conditions on venous ECs while it was flow-dependent on arterial cells. Furthermore, analysis of HS/glycocalyx response after stimulation with inflammatory cues revealed that venous, but not arterial ECs, are resistant to HS shedding. This finding was observed also on isolated porcine vessels. Persistence of HS on venous ECs prevented complement deposition and clot formation after stimulation with tumor necrosis factor α or lipopolysaccharide, whereas after xenogeneic activation no glycocalyx-mediated protection was observed. Contrarily, HS shedding on arterial cells, even without an inflammatory insult, was sufficient to induce a proinflammatory and procoagulant phenotype. Our data indicate that the dimorphic response of arterial and venous ECs is partially due to distinct HS/glycocalyx dynamics suggesting that arterial and venous thrombo-inflammatory disorders require targeted therapies.


Asunto(s)
Células Endoteliales , Heparitina Sulfato , Animales , Porcinos , Células Endoteliales/metabolismo , Heparitina Sulfato/metabolismo , Glicocálix/metabolismo , Arterias/metabolismo , Venas/metabolismo
6.
Development ; 150(7)2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36938965

RESUMEN

Blood vessels form elaborate networks that depend on tissue-specific signalling pathways and anatomical structures to guide their growth. However, it is not clear which morphogenetic principles organize the stepwise assembly of the vasculature. We therefore performed a longitudinal analysis of zebrafish caudal fin vascular assembly, revealing the existence of temporally and spatially distinct morphogenetic processes. Initially, vein-derived endothelial cells (ECs) generated arteries in a reiterative process requiring vascular endothelial growth factor (Vegf), Notch and cxcr4a signalling. Subsequently, veins produced veins in more proximal fin regions, transforming pre-existing artery-vein loops into a three-vessel pattern consisting of an artery and two veins. A distinct set of vascular plexuses formed at the base of the fin. They differed in their diameter, flow magnitude and marker gene expression. At later stages, intussusceptive angiogenesis occurred from veins in distal fin regions. In proximal fin regions, we observed new vein sprouts crossing the inter-ray tissue through sprouting angiogenesis. Together, our results reveal a surprising diversity among the mechanisms generating the mature fin vasculature and suggest that these might be driven by separate local cues.


Asunto(s)
Células Endoteliales , Pez Cebra , Animales , Pez Cebra/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Neovascularización Fisiológica , Venas/metabolismo
7.
Cell Mol Life Sci ; 79(9): 491, 2022 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-35987946

RESUMEN

Endothelial cells in veins differ in morphology, function and gene expression from those in arteries and lymphatics. Understanding how venous and arterial identities are induced during development is required to understand how arterio-venous malformations occur, and to improve the outcome of vein grafts in surgery by promoting arterialization of veins. To identify factors that promote venous endothelial cell fate in vivo, we isolated veins from quail embryos, at different developmental stages, that were grafted into the coelom of chick embryos. Endothelial cells migrated out from the grafted vein and their colonization of host veins and/or arteries was quantified. We show that venous fate is promoted by sympathetic vessel innervation at embryonic day 11. Removal of sympathetic innervation decreased vein colonization, while norepinephrine enhanced venous colonization. BMP treatment or inhibition of ERK enhanced venous fate, revealing environmental neurotransmitter and BMP signaling and intrinsic ERK inhibition as actors in venous fate acquisition. We also identify the BMP antagonist Noggin as a potent mediator of venous arterialization.


Asunto(s)
Células Endoteliales , Venas , Animales , Arterias , Diferenciación Celular/fisiología , Embrión de Pollo , Transducción de Señal , Venas/metabolismo
8.
J Vasc Surg Venous Lymphat Disord ; 10(2): 469-481.e2, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34506963

RESUMEN

OBJECTIVE: Venous malformations (VMs) are the most frequent vascular malformations and are characterized by dilated and tortuous veins with a dysregulated vascular extracellular matrix. The purpose of the present study was to investigate the potential involvement of microRNA-21 (miR-21), a multifunctional microRNA tightly associated with extracellular matrix regulation, in the pathogenesis of VMs. METHODS: The expression of miR-21, collagen I, III, and IV, transforming growth factor-ß (TGF-ß), and Smad3 (mothers against decapentaplegic homolog 3) was evaluated in VMs and normal skin tissue using in situ hybridization, immunohistochemistry, Masson trichrome staining, and real-time polymerase chain reaction. Human umbilical vein endothelial cells (HUVECs) were used to explore the underlying mechanisms. RESULTS: miR-21 expression was markedly decreased in the VM specimens compared with normal skin, in parallel with downregulation of collagen I, III, and IV and the TGF-ß/Smad3 pathway in VMs. Moreover, our data demonstrated that miR-21 positively regulated the expression of collagens in HUVECs and showed a positive association with the TGF-ß/Smad3 pathway in the VM tissues. In addition, miR-21 was found to mediate TGF-ß-induced upregulation of collagens in HUVECs. Our data have indicated that miR-21 and the TGF-ß/Smad3 pathway could form a positive feedback loop to synergistically regulate endothelial collagen synthesis. In addition, TGF-ß/Smad3/miR-21 feedback loop signaling was upregulated in bleomycin-treated HUVECs and VM specimens, which was accompanied by increased collagen deposition. CONCLUSIONS: To the best of our knowledge, the present study has, for the first time, revealed downregulation of miR-21 in VMs, which might contribute to decreased collagen expression via the TGF-ß/Smad3/miR-21 signaling feedback loop. These findings provide new information on the pathogenesis of VMs and might facilitate the development of new therapies for VMs.


Asunto(s)
Colágeno/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , MicroARNs/metabolismo , Piel/irrigación sanguínea , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Malformaciones Vasculares/metabolismo , Venas/metabolismo , Bleomicina/toxicidad , Estudios de Casos y Controles , Células Cultivadas , Colágeno/genética , Regulación hacia Abajo , Retroalimentación Fisiológica , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , MicroARNs/genética , Fosforilación , Transducción de Señal , Malformaciones Vasculares/genética , Malformaciones Vasculares/patología , Venas/anomalías , Venas/patología
9.
Development ; 149(3)2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34931661

RESUMEN

Endothelial cell migration and proliferation are essential for the establishment of a hierarchical organization of blood vessels and optimal distribution of blood. However, how these cellular processes are quantitatively coordinated to drive vascular network morphogenesis remains unknown. Here, using the zebrafish vasculature as a model system, we demonstrate that the balanced distribution of endothelial cells, as well as the resulting regularity of vessel calibre, is a result of cell migration from veins towards arteries and cell proliferation in veins. We identify the Wiskott-Aldrich Syndrome protein (WASp) as an important molecular regulator of this process and show that loss of coordinated migration from veins to arteries upon wasb depletion results in aberrant vessel morphology and the formation of persistent arteriovenous shunts. We demonstrate that WASp achieves its function through the coordination of junctional actin assembly and PECAM1 recruitment and provide evidence that this is conserved in humans. Overall, we demonstrate that functional vascular patterning in the zebrafish trunk is established through differential cell migration regulated by junctional actin, and that interruption of differential migration may represent a pathomechanism in vascular malformations.


Asunto(s)
Vasos Sanguíneos/crecimiento & desarrollo , Morfogénesis/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Proteína del Síndrome de Wiskott-Aldrich/genética , Actinas/genética , Animales , Arterias/crecimiento & desarrollo , Arterias/metabolismo , Movimiento Celular/genética , Proliferación Celular/genética , Células Endoteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Uniones Intercelulares/genética , Venas/crecimiento & desarrollo , Venas/metabolismo , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo
10.
PLoS One ; 16(12): e0261073, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34905581

RESUMEN

Patients with chronic venous disease (CVD) have elevated levels of leucocyte elastase (LE) released from the activation of leucocytes. In acute deep venous thrombosis (DVT), LE can degrade fibrin from the thrombus resulting in cross-linked fibrin degradation products (E-XDP) being released into the bloodstream. In patients with CVD the levels and significance of circulating E-XDP are unknown. We aimed to investigate the association between plasma E-XDP concentration and severity of CVD. Levels of E-XDP were quantified with a specific enzyme-linked immunosorbent assay (ELISA) in plasma from 142 consecutively recruited CVD patients (mean age 64 years, (range 23-89), 81 were females and 61 males). Patients were also divided into three groups based on CVD severity using the C-class of the Clinical-Etiological-Anatomical-Pathophysiological (CEAP) classification, with C 0-1 class as the reference group, C 2-3 as the second group and C 4-6 as the third group with the most severely affected patients. We found significantly elevated levels of E-XDP in patients with C 4-6 compared with patients with C 0-1 (p = 0.007) and increased with increasing disease severity across the groups (p = 0.02). Significant independent association was observed between levels of E-XDP and the classes C 4-6 after adjustment for age and sex (p < 0.05), but the association was no longer significant after further adjustment for use of statins, use of anticoagulants and history of DVT (p = 0.247). This exploratory study shows that E-XDP levels are elevated in patients with CVD, encouraging further studies on the role of E-XDP in CVD.


Asunto(s)
Productos de Degradación de Fibrina-Fibrinógeno/análisis , Elastasa de Leucocito/metabolismo , Enfermedades Vasculares/patología , Venas , Adulto , Anciano , Anciano de 80 o más Años , Enfermedad Crónica , Femenino , Fibrina/metabolismo , Fibrinólisis , Humanos , Masculino , Persona de Mediana Edad , Venas/metabolismo , Venas/patología , Adulto Joven
11.
Adv Sci (Weinh) ; 8(23): e2101837, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34693660

RESUMEN

Neurovascular disorders, which involve the vascular and nervous systems, are common. Research on such disorders usually focuses on either vascular or nervous components, without looking at how they interact. Adopting a neurovascular perspective is essential to improve current treatments. Therefore, comparing molecular processes known to be involved in both systems separately can provide insight into promising areas of future research. Since development and regeneration share many mechanisms, comparing signaling molecules involved in both the developing vascular and nervous systems and shedding light to those that they have in common can reveal processes, which have not yet been studied from a regenerative perspective, yet hold great potential. Hence, this review discusses and compares processes involved in the development of the vascular and nervous systems, in order to provide an overview of the molecular mechanisms, which are most promising with regards to treatment for neurovascular disorders. Vascular endothelial growth factor, semaphorins, and ephrins are found to hold the most potential, while fibroblast growth factor, bone morphogenic protein, slits, and sonic hedgehog are shown to participate in both the developing vascular and nervous systems, yet have not been studied at the neurovascular level, therefore being of special interest for future research.


Asunto(s)
Arterias/embriología , Sistema Nervioso/embriología , Medicina Regenerativa/métodos , Transducción de Señal , Venas/embriología , Arterias/metabolismo , Efrinas/metabolismo , Femenino , Factores de Crecimiento de Fibroblastos/metabolismo , Humanos , Sistema Nervioso/metabolismo , Semaforinas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Venas/metabolismo
12.
Cell Rep ; 36(2): 109346, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34260918

RESUMEN

The spleen comprises defined microanatomical compartments that uniquely contribute to its diverse host defense functions. Here, we identify a vascular compartment within the red pulp of the spleen delineated by expression of the atypical chemokine receptor 4 (ACKR4) in endothelial cells. ACKR4-positive vessels form a three-dimensional sinusoidal network that connects via shunts to the marginal sinus and tightly surrounds the outer perimeter of the marginal zone. Endothelial cells lining this vascular compartment express ACKR4 as part of a distinct gene expression profile. We show that T cells enter the spleen largely through this peri-marginal sinus and initially localize extravascularly around these vessels. In the absence of ACKR4, homing of T cells into the spleen and subsequent migration into T cell areas is impaired, and organization of the marginal zone is severely affected. Our data delineate the splenic peri-marginal sinus as a compartment that supports spleen homing of T cells.


Asunto(s)
Receptores CCR/metabolismo , Bazo/irrigación sanguínea , Animales , Animales Recién Nacidos , Circulación Sanguínea , Movimiento Celular , Quimiocina CCL19/metabolismo , Células Endoteliales/metabolismo , Vasos Linfáticos/metabolismo , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T , Venas/metabolismo
13.
Elife ; 102021 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-34227466

RESUMEN

Pericytes have been implicated in various neuropathologies, yet little is known about their function and signaling pathways in health. Here, we characterized calcium dynamics of cortical mural cells in anesthetized or awake Pdgfrb-CreERT2;Rosa26< LSL-GCaMP6s > mice and in acute brain slices. Smooth muscle cells (SMCs) and ensheathing pericytes (EPs), also named as terminal vascular SMCs, revealed similar calcium dynamics in vivo. In contrast, calcium signals in capillary pericytes (CPs) were irregular, higher in frequency, and occurred in cellular microdomains. In the absence of the vessel constricting agent U46619 in acute slices, SMCs and EPs revealed only sparse calcium signals, whereas CPs retained their spontaneous calcium activity. Interestingly, chemogenetic activation of neurons in vivo and acute elevations of extracellular potassium in brain slices strongly decreased calcium activity in CPs. We propose that neuronal activation and an extracellular increase in potassium suppress calcium activity in CPs, likely mediated by Kir2.2 and KATP channels.


Asunto(s)
Encéfalo/metabolismo , Señalización del Calcio/fisiología , Calcio/metabolismo , Animales , Encéfalo/patología , Capilares/metabolismo , Femenino , Masculino , Ratones , Músculo Liso Vascular/diagnóstico por imagen , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Pericitos/citología , Pericitos/fisiología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Vasoconstricción , Venas/metabolismo
14.
Aging (Albany NY) ; 13(12): 16656-16666, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-34170847

RESUMEN

Venous neointimal hyperplasia can be a problem after vein interventions. We hypothesized that inhibiting programmed death-1 (PD-1) can decrease venous neointimal hyperplasia in a rat inferior vena cava (IVC) patch venoplasty model. The rats were divided into four groups: the control group was only decellularized without other special treatment; the PD-1 group was injected with a single dose of humanized PD-1 antibody (4 mg/kg); the PD-1 antibody coated patches group; the BMS-1 (a PD-1 small molecular inhibitor) coated patches group (PD-1 inhibitor-1). Patches were implanted to the rat IVC and harvested on day 14 and analyzed. Immunohistochemical analysis showed PD-1-positive cells in the neointima in the human samples. There was high protein expression of PD-1 in the neointima in the rat IVC venoplasty model. PD-1 antibody injection can significantly decrease neointimal thickness (p < 0.0001). PD-1 antibody or BMS-1 was successfully conjugated to the decellularized rat thoracic artery patch by hyaluronic acid with altered morphology and reduced the water contact angle (WCA). Patches coated with humanized PD-1 antibody or BMS-1 both can also decrease neointimal hyperplasia and inflammatory cells infiltration. PD-1-positive cells are present in venous neointima in both human and rat samples. Inhibition of the PD-1 pathway may be a promising therapeutic strategy to inhibit venous neointimal hyperplasia.


Asunto(s)
Neointima/metabolismo , Neointima/patología , Receptor de Muerte Celular Programada 1/metabolismo , Venas/metabolismo , Venas/patología , Animales , Humanos , Hiperplasia/metabolismo , Inyecciones Intraperitoneales , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Ratas , Bibliotecas de Moléculas Pequeñas/farmacología , Agua
15.
Horm Metab Res ; 53(6): 364-370, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34154027

RESUMEN

The aim of the study was to investigate the significance and influence of adrenocorticotropic hormone (ACTH) stimulation in primary aldosteronism (PA) patients with simultaneous bilateral adrenal vein sampling (AVS). All patients diagnosed with PA underwent simultaneous bilateral AVS with ACTH. In 95 patients, the post-ACTH SI significantly increased (p<0.001), and it gradually decreased from t10-t30 after ACTH stimulation (p<0.001). The unsuccessful catheterization decreased after ACTH stimulation. Time points within 20 min after ACTH stimulation were better for sampling, and the selectivity did not increase over longer periods. According to lateralization before and after ACTH stimulation, the patients could be divided into 3 groups (U, unilateral; B, bilateral): U/U , U/B or B/U, and B/B. Compared with the U/U group, in the U/B or B/U and B/B groups, the lateralization index (LI) was lower both at baseline and after ACTH stimulation (p<0.0001), the contralateral index (CLI) was higher after ACTH stimulation (p<0.003), the serum potassium level was higher (p<0.001), and the carbon dioxide combining power (CO2CP) and base excess (BE) levels were lower. In conclusion, in simultaneous bilateral AVS, ACTH stimulation had significant effects on increasing the catheterization selectivity. Lateralization change was observed after stimulation. After ACTH stimulation, fewer patients could be diagnosed with lateralized PA. Patients with consistent lateralized PA showed a more serious phenotype.


Asunto(s)
Glándulas Suprarrenales/irrigación sanguínea , Hormona Adrenocorticotrópica/farmacología , Biomarcadores/sangre , Recolección de Muestras de Sangre/métodos , Hiperaldosteronismo/diagnóstico , Venas/metabolismo , Glándulas Suprarrenales/efectos de los fármacos , Adulto , Anciano , Cateterismo , Femenino , Estudios de Seguimiento , Humanos , Hiperaldosteronismo/sangre , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Prospectivos , Venas/efectos de los fármacos , Adulto Joven
16.
BMC Endocr Disord ; 21(1): 114, 2021 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-34059026

RESUMEN

BACKGROUND: Primary aldosteronism (PA) is highly prevalent in hypertensive population. Adrenal vein sampling (AVS) is the only procedure to assess adrenal aldosterone hypersecretion in PA. PA patients without aldosterone-producing adenomas (APA) frequently have unilateral aldosterone hypersecretion (UAH). These patients could bear inappropriate adrenalectomy without AVS. This study aims to identify which clinical characteristics should be recommended to perform AVS in these PA patients. METHODS: This study was performed from January 2018 to July 2019 at a center for hypertension and metabolic diseases. Adrenal computed tomography (CT) scan, biochemical evaluation, and AVS were performed. RESULTS: Total 141 patients were included in this study. Aldosterone to renin ratio (ARR) after confirmatory test is highly associated with adrenal laterality. The specificity of ARR > 10 (ng/dL)/(mU/L) after confirmatory test is 100%. After confirmatory test, patients with ARR > 10 (ng/dL)/(mU/L) had higher plasma aldosterone concentration and incidences of ischemic heart diseases and renal damage(p < 0.05). CONCLUSIONS: After confirmatory tests, ARR > 10 (ng/dL)/(mU/L) indicates adrenal laterality, with increasingly cardiorenal damage in PA patients without APA. Thus, AVS should be recommended in these patients before surgery. TRIAL REGISTRATION: NCT03398785 , Date of Registration: December 24, 2017.


Asunto(s)
Glándulas Suprarrenales/metabolismo , Adenoma Corticosuprarrenal/patología , Aldosterona/metabolismo , Hiperaldosteronismo/diagnóstico , Hipertensión/complicaciones , Venas/metabolismo , Adenoma Corticosuprarrenal/metabolismo , Diagnóstico Diferencial , Femenino , Estudios de Seguimiento , Humanos , Hiperaldosteronismo/etiología , Hiperaldosteronismo/metabolismo , Masculino , Persona de Mediana Edad , Pronóstico , Tomografía Computarizada por Rayos X
18.
Biomed Chromatogr ; 35(7): e5089, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33569806

RESUMEN

An arterialized venous flap (AVF) is an ideal choice of flap to repair wounds. However, the survival of these flaps remains the source of some concern. This study used metabolomic analysis to investigate the mechanisms underlying survival in AVF flaps in order to guide the clinical application of these flaps. Thirty-six male Japanese rabbits were randomly divided into a sham group and an AVF group. They were used for histology and hemodynamic investigations. Three days after surgery, tissue samples were analyzed by mass spectroscopy-based metabolomics. The results of the study revealed a reduction in blood flow, infiltration of inflammatory cells, and necrosis of flaps in the AVF group. In addition, notable changes were evident in the levels of several metabolites in the AVF group, including lactic acid, acetoacetic acid, inositol phosphate, arachidonic acid, and other metabolites. Our results indicate that the AVF group experienced changes in several biological pathways, including energy metabolism, cell membrane stability, and inflammatory response. There is a significant metabolic difference between AVFs and physiological flaps. The dysregulation in certain metabolites may be related to the specific hemodynamics and insufficient energy metabolism of the AVFs.


Asunto(s)
Arterias , Espectrometría de Masas/métodos , Metabolómica/métodos , Colgajos Quirúrgicos/irrigación sanguínea , Venas , Animales , Arterias/química , Arterias/metabolismo , Masculino , Metaboloma/fisiología , Conejos , Venas/química , Venas/metabolismo
19.
Angiogenesis ; 24(2): 251-269, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33449300

RESUMEN

Endothelial cells display an extraordinary plasticity both during development and throughout adult life. During early development, endothelial cells assume arterial, venous, or lymphatic identity, while selected endothelial cells undergo additional fate changes to become hematopoietic progenitor, cardiac valve, and other cell types. Adult endothelial cells are some of the longest-lived cells in the body and their participation as stable components of the vascular wall is critical for the proper function of both the circulatory and lymphatic systems, yet these cells also display a remarkable capacity to undergo changes in their differentiated identity during injury, disease, and even normal physiological changes in the vasculature. Here, we discuss how endothelial cells become specified during development as arterial, venous, or lymphatic endothelial cells or convert into hematopoietic stem and progenitor cells or cardiac valve cells. We compare findings from in vitro and in vivo studies with a focus on the zebrafish as a valuable model for exploring the signaling pathways and environmental cues that drive these transitions. We also discuss how endothelial plasticity can aid in revascularization and repair of tissue after damage- but may have detrimental consequences under disease conditions. By better understanding endothelial plasticity and the mechanisms underlying endothelial fate transitions, we can begin to explore new therapeutic avenues.


Asunto(s)
Diferenciación Celular , Células Endoteliales/metabolismo , Neovascularización Fisiológica , Heridas y Lesiones/metabolismo , Pez Cebra/metabolismo , Animales , Arterias/metabolismo , Células Madre Hematopoyéticas , Humanos , Vasos Linfáticos/metabolismo , Venas/metabolismo , Heridas y Lesiones/terapia
20.
Dev Biol ; 473: 1-14, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33453264

RESUMEN

Correct vascular differentiation requires distinct patterns of gene expression in different subtypes of endothelial cells. Members of the ETS transcription factor family are essential for the transcriptional activation of arterial and angiogenesis-specific gene regulatory elements, leading to the hypothesis that they play lineage-defining roles in arterial and angiogenic differentiation directly downstream of VEGFA signalling. However, an alternative explanation is that ETS binding at enhancers and promoters is a general requirement for activation of many endothelial genes regardless of expression pattern, with subtype-specificity provided by additional factors. Here we use analysis of Ephb4 and Coup-TFII (Nr2f2) vein-specific enhancers to demonstrate that ETS factors are equally essential for vein, arterial and angiogenic-specific enhancer activity patterns. Further, we show that ETS factor binding at these vein-specific enhancers is enriched by VEGFA signalling, similar to that seen at arterial and angiogenic enhancers. However, while arterial and angiogenic enhancers can be activated by VEGFA in vivo, the Ephb4 and Coup-TFII venous enhancers are not, suggesting that the specificity of VEGFA-induced arterial and angiogenic enhancer activity occurs via non-ETS transcription factors. These results support a model in which ETS factors are not the primary regulators of specific patterns of gene expression in different endothelial subtypes.


Asunto(s)
Células Endoteliales/metabolismo , Neovascularización Fisiológica/fisiología , Proteínas Proto-Oncogénicas c-ets/metabolismo , Animales , Arterias/metabolismo , Diferenciación Celular/fisiología , Células Endoteliales/fisiología , Endotelio/metabolismo , Elementos de Facilitación Genéticos/genética , Femenino , Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-ets/fisiología , Transducción de Señal , Factores de Transcripción/metabolismo , Activación Transcripcional , Factor A de Crecimiento Endotelial Vascular/metabolismo , Venas/metabolismo , Pez Cebra/embriología , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA