Добірка наукової літератури з теми "Angiogenesis, Autophagy, ROP"

Оформте джерело за APA, MLA, Chicago, Harvard та іншими стилями

Оберіть тип джерела:

Ознайомтеся зі списками актуальних статей, книг, дисертацій, тез та інших наукових джерел на тему "Angiogenesis, Autophagy, ROP".

Біля кожної праці в переліку літератури доступна кнопка «Додати до бібліографії». Скористайтеся нею – і ми автоматично оформимо бібліографічне посилання на обрану працю в потрібному вам стилі цитування: APA, MLA, «Гарвард», «Чикаго», «Ванкувер» тощо.

Також ви можете завантажити повний текст наукової публікації у форматі «.pdf» та прочитати онлайн анотацію до роботи, якщо відповідні параметри наявні в метаданих.

Статті в журналах з теми "Angiogenesis, Autophagy, ROP"

1

Ash, Dipankar, Sudhahar Varadarajan, Seock-Won Youn, Mustafa N. Okur, John P. O'Brien, Malgorzata McMenamin, Xuexiu Fang, et al. "Abstract 13610: Endothelial Cu Transporter Atp7a Promotes Vegfr2 Signaling and Post-ischemic Neovascularization via Regulating Autophagy." Circulation 142, Suppl_3 (November 17, 2020). http://dx.doi.org/10.1161/circ.142.suppl_3.13610.

Повний текст джерела
Анотація:
Background: VEGFR2 (KDR/Flk1) signaling in endothelial cells (ECs) plays a central role in angiogenesis. Copper (Cu) is essential micronutrient and has been implicated in angiogenesis. The P-type ATPase transporter ATP7A is key regulator of Cu homeostasis but its role in VEGFR2 signaling in ECs and post-ischemic neovascularization is entirely unknown. Results: Here we show that ATP7A expression was dramatically increased in the angiogenic ECs in mice hindlimb ischemia model. EC-specific ATP7A deficient mice or Cu transporter-dysfunctional ATP7A mut mice showed significant decrease in blood flow recovery and CD31+ capillary density (angiogenesis) in ischemic tissues compared to their control mice. In cultured human ECs, ATP7A knockdown with siRNA significantly inhibited VEGF-induced migration (67%), capillary network formation on Matrigel (46%). Immunofluorescence, co-immunoprecipitation, and proximity ligation assays showed that VEGF stimulated ATP7A translocation from the trans-Golgi network to the plasma membrane where it bound to VEGFR2. Surprisingly, loss of ATP7A promoted VEGF-induced VEGFR2 protein degradation (56%) and inhibited VEGFR2 signaling via enhancing VEGFR2 ubiquitination (2.0-fold) in a Cu-independent manner. This was associated with reduced cell surface VEGFR2 expression, increased VEGFR2 binding to selective autophagic cargo/adaptor p62/SQSTM1 (60%) and LC3-GFP+autophagic puncta (34%) and autolysosome formation (transmission electron microscopy). Inhibition of autophagy by bafilomycin A1 or chloroquine prevented enhanced VEGFR2 degradation in ATP7A-depleted ECs. Furthermore, overexpression of p62, but not p62 lacking ubiquitin binding domain, or autophagy inducer rapamycin treatment increased VEGFR2 degradation (26% and 87%, respectively) and p62 overexpression inhibited ECs migration (29%). Enhanced autophagy flux due to ATP7A dysfunction in vivo was confirmed by employing autophagy reporter CAG-ATP7A mut -RFP-EGFP-LC3 transgenic mice. Conclusion: Our study uncovers the unexpected and novel function of ATP7A to limit autophagic degradation of VEGFR2, thereby promoting VEGFR2 signaling and angiogenesis, which restores neovascularization in ischemic disease.
Стилі APA, Harvard, Vancouver, ISO та ін.
2

Ash, Dipankar, Varadarajan Sudhahar, Seock-Won Youn, Mustafa Nazir Okur, Archita Das, John P. O’Bryan, Maggie McMenamin, et al. "The P-type ATPase transporter ATP7A promotes angiogenesis by limiting autophagic degradation of VEGFR2." Nature Communications 12, no. 1 (May 25, 2021). http://dx.doi.org/10.1038/s41467-021-23408-1.

Повний текст джерела
Анотація:
AbstractVEGFR2 (KDR/Flk1) signaling in endothelial cells (ECs) plays a central role in angiogenesis. The P-type ATPase transporter ATP7A regulates copper homeostasis, and its role in VEGFR2 signaling and angiogenesis is entirely unknown. Here, we describe the unexpected crosstalk between the Copper transporter ATP7A, autophagy, and VEGFR2 degradation. The functional significance of this Copper transporter was demonstrated by the finding that inducible EC-specific ATP7A deficient mice or ATP7A-dysfunctional ATP7Amut mice showed impaired post-ischemic neovascularization. In ECs, loss of ATP7A inhibited VEGF-induced VEGFR2 signaling and angiogenic responses, in part by promoting ligand-induced VEGFR2 protein degradation. Mechanistically, VEGF stimulated ATP7A translocation from the trans-Golgi network to the plasma membrane where it bound to VEGFR2, which prevented autophagy-mediated lysosomal VEGFR2 degradation by inhibiting autophagic cargo/adapter p62/SQSTM1 binding to ubiquitinated VEGFR2. Enhanced autophagy flux due to ATP7A dysfunction in vivo was confirmed by autophagy reporter CAG-ATP7Amut -RFP-EGFP-LC3 transgenic mice. In summary, our study uncovers a novel function of ATP7A to limit autophagy-mediated degradation of VEGFR2, thereby promoting VEGFR2 signaling and angiogenesis, which restores perfusion recovery and neovascularization. Thus, endothelial ATP7A is identified as a potential therapeutic target for treatment of ischemic cardiovascular diseases.
Стилі APA, Harvard, Vancouver, ISO та ін.
3

Li, Baolong, Zhengtai Chen, Xiaobin Luo, Chenxi Zhang, Hongyu Chen, Shuxuan Wang, Mengyao Zhao, et al. "Butylphthalide Inhibits Autophagy and Promotes Multiterritory Perforator Flap Survival." Frontiers in Pharmacology 11 (January 29, 2021). http://dx.doi.org/10.3389/fphar.2020.612932.

Повний текст джерела
Анотація:
Multiterritory perforator flap is an important plastic surgery technique, yet its efficacy can be limited by partial necrosis at the choke Ⅱ zone. Butylphthalide (NBP) has been used for many diseases but has not been studied in the multiterritory perforator flap. With the effect of NBP, we observed increasing in capillary density, inhibition of autophagy and oxidative stress, and a reduction in apoptosis of cells, all consistent with increased flap survival. However, the protective effect of NBP on multiterritory perforator flap was lost following administration of the autophagy agonist rapamycin (Rap). Through the above results, we assumed that NBP promotes flap survival by inhibiting autophagy. Thus, this study has found a new pharmacological effect of NBP on the multiterritory perforator by inhibiting autophagy to prevent distal postoperative necrosis and exert effects on angiogenesis, oxidative stress, and apoptosis within the flap.
Стилі APA, Harvard, Vancouver, ISO та ін.
4

Xue, Lianguo, Tao Jia, Yuanxin Zhu, Lidong Zhao, and Jianping Mao. "Down-regulation of circ_0058058 suppresses proliferation, angiogenesis and metastasis in multiple myeloma through miR-338-3p/ATG14 pathway." Journal of Orthopaedic Surgery and Research 16, no. 1 (December 2021). http://dx.doi.org/10.1186/s13018-021-02867-8.

Повний текст джерела
Анотація:
Abstract Background Multiple myeloma (MM) is one of the most frequently diagnosed hematological malignancy. Dysregulation of circular RNAs (circRNAs) has important impacts on MM process. Herein, this work aimed to investigate the role and mechanism of circ_0058058 in MM progression. Methods Levels of genes and proteins were detected by real-time reverse transcription PCR (RT-qPCR) and Western blot. CCK-8 assay, colony formation assay, EdU assay, flow cytometry, tube formation assay, transwell assay and Western blot were utilized to detect the proliferation, apoptosis, angiogenesis and metastasis of MM cells. The target relationship between miR-338-3p and circ_0058058 or ATG14 (autophagy related 14) was verified by dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay. In vivo experiments were performed using Xenograft assay. Results Circ_0058058 was up-regulated in MM bone marrow aspirates and cells, knockdown of circ_0058058 reduced MM cell proliferation, angiogenesis and metastasis, but induced apoptosis in vitro. In a MM xenograft mouse model, circ_0058058 silencing reduced MM tumor growth and cell proliferation. Mechanistically, circ_0058058 acted as a sponge for miR-338-3p to up-regulate ATG14 expression, which was validated to be a target of miR-338-3p. Rescue assay showed that miR-338-3p inhibition reversed the antitumor effects of circ_0058058 knockdown on MM cell. Moreover, forced expression of miR-338-3p suppressed MM cell malignant phenotype, which was abolished by ATG14 up-regulation. Conclusion Circ_0058058 functions as a sponge for miR-338-3p to elevate ATG14 expression to promote MM cell proliferation, metastasis and angiogenesis, affording a potential therapeutic target for MM prevention.
Стилі APA, Harvard, Vancouver, ISO та ін.

Дисертації з теми "Angiogenesis, Autophagy, ROP"

1

Pesce, Noemi Anna. "MOLECULAR MECHANISMS OF ANGIOGENESIS-RELATED OCULAR DISEASES IN PRECLINICAL MODELS." Doctoral thesis, Università di Siena, 2021. http://hdl.handle.net/11365/1148688.

Повний текст джерела
Анотація:
Neovascularization in the eye contributes to visual loss in several ocular diseases, including proliferative diabetic retinopathy (PDR), neovascular glaucoma (NVG) and retinopathy of prematurity (ROP). In these diseases, the neovascular mechanisms originate from the retina, but some advanced stages of PDR or nG can lead to the development of rubeosis iridis (RI), a clinical manifestation characterized by iris angiogenesis. Vascular endothelial growth factors (VEGFs) and their receptor (VEGFRs) are the key promoters of angiogenesis, playing a crucial role in both physiological and pathological angiogenesis. For this reason, anti-VEGF therapies have represented a great step forward in the treatment of ocular neovascular diseases, but the effects can be somewhat limited. In addition, beyond the vascular alterations, degenerative changes might occur in the neuroretina, including inflammation and neurodegeneration, which further compromise visual function in people affected by ocular diseases. Consequently, a pharmacologic strategy aimed at inhibiting several pathways might be a more suitable therapeutic approach. A first suitable target may be represented by the pathway triggered by the urokinase plasminogen activator (uPA) and its receptor (uPAR), which play a pivotal role in extracellular matrix remodeling during angiogenesis, where uPAR, a glycosylphosphatidylinositol-anchored protein, regulates cell migration and proliferation through the assembly with transmembrane receptors. An additional pathway that may represent an interesting target is autophagy, as an expanding body of literature suggests that mechanisms of autophagy are involved in neurodegeneration in ocular disease. Autophagy is an essential process in maintaining the normal cellular homeostasis and energy balance. In retinal diseases, autophagy contributes to retinal cells protecting themselves against harmful stress stimuli; however, dysregulated autophagy may result in retinal deterioration. Here, we investigate the effects of the uPAR antagonist—UPARANT—on the angiogenic and inflammatory processes, in a context of hypoxia-induced angiogenesis, using a novel ex vivo human iris angiogenesis assay and, an in vivo mouse model of RI-associated with proliferative retinopathy (PR). A rat model of oxygen-induced retinopathy (OIR), an acknowledged model of ROP, was used to evaluate neuroretinal changes in the expression of key mediators of autophagy, induced by disease. First, we investigated the autophagic profile in newborn rats and, subsequently determined a correlation between autophagy and mechanisms of cell death in the OIR rat retina. In addition, an inhibitor of autophagy was valuated to analyze the possible effects of in mitigating autophagy-associated cell death and retinal function in the OIR rat model.
Стилі APA, Harvard, Vancouver, ISO та ін.
Ми пропонуємо знижки на всі преміум-плани для авторів, чиї праці увійшли до тематичних добірок літератури. Зв'яжіться з нами, щоб отримати унікальний промокод!

До бібліографії