Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Langmuir ; 40(19): 10024-10034, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38698547

RESUMO

Responsive Pickering emulsions, with unique nanoparticle interfaces and sensitivity to external stimuli, significantly enhanced the stability and applicability of Pickering emulsions. Multifunctional composite material poly((2-(dimethylaminoethyl methacrylate)-b-(acrylate cyclodextrin))/Fe3O4 nanoparticles, namely P(DMAEMA-b-A-CD)/Fe3O4, with both multiresponsive characteristics and emulsifying capabilities had been designed to remove small oil droplets from water. Using the reversible addition-fragmentation chain transfer (RAFT) method, diblock polymers P(DMAEMA-b-A-CD) were grown in a controlled manner on the surface of Fe3O4. The Fe3O4 core showed responsiveness to a magnetic field, and the block copolymers prepared via the RAFT method demonstrated reactivity to both pH and CO2. The P(DMAEMA-b-A-CD)/Fe3O4 nanoparticles exhibited the capability to form Pickering/Oxford emulsions with exceptional stabilization properties. It could be observed that the introduction of CO2, acid, and a magnetic field led to the breakage of the emulsion, while the emulsion could be restabilized by removing the CO2 and the magnetic field or by adding alkali. Measurements of interfacial tension, ζ-potential, and contact angle demonstrated that the emulsification/breakdown mechanisms associated with pH and CO2/N2 were related to the surface wettability of the nanoparticles. In addition, the emulsifier had an excellent cycling capacity with at least 10 cycles by CO2/N2. Additionally, P(DMAEMA-b-A-CD)/Fe3O4 nanoparticles exhibited excellent stability in oil phases with large polarity differences and various real oil phases with different viscosities. Importantly, the P(DMAEMA-b-A-CD)/Fe3O4 nanoparticles could serve as functional materials for efficiently separating small oil droplets from water through the application of a magnetic field. Therefore, P(DMAEMA-b-A-CD)/Fe3O4 nanoparticles held promising potential as materials with economic and commercial value for oil-water separation applications.

2.
Heliyon ; 10(8): e29504, 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38655349

RESUMO

Despite growing evidence suggesting an important contribution of Tumor Protein P53 Inducible Protein 11 (TP53I11) in cancer progression, the role of TP53I11 remains unclear. Our first pan-cancer analysis of TP53I11 showed some tumor tissues displayed reduced TP53I11 expression compared to normal tissues, while others exhibited high TP53I11 expression. Meanwhile, TP53I11 expression carries a particular pan-cancer risk, as high TP53I11 expression levels are detrimental to survival for BRCA, KIRP, MESO, and UVM, but to beneficial survival for KIRC. We demonstrated that TP53I11 expression negatively correlates with DNA methylation in most cancers, and the S14 residue of TP53I11 is phosphorylated in several cancer types. Additionally, TP53I11 was found to be associated with endothelial cells in pan-cancer, and functional enrichment analysis provided strong evidence for its role in tumor angiogenesis. In vitro angiogenesis assays confirmed that TP53I11 can promote angiogenic function of human umbilical vein endothelial cells (HUVECs) in vitro. Mechanistic investigations reveal that TP53I11 is transcriptionally up-regulated by HIF2A under hypoxia.

3.
ACS Appl Mater Interfaces ; 16(14): 17715-17727, 2024 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-38551105

RESUMO

To ensure safety and efficiency in the production and transportation of fuel oil, there is an urgent demand to develop intelligent emulsifiers to deal with this challenge. Fe3O4@PDA-P(NIPAM-b-MAA-b-LMA) (MNPDNML) microspheres were prepared by modifying polydopamine and the triblock polymer brush P(NIPAM-b-MAA-b-LMA) on the surface of Fe3O4 nanoparticles via oxidative autopolymerization and SI-RAFT polymerization. Therefore, the MNPDNML microspheres exhibited sensitive stimulus-responsive behavior to pH, temperature, near-infrared (NIR) laser radiation, and magnetic fields. The stability state of the emulsion could be modulated by changing pH, temperature, magnetic field, and NIR radiation, and the reversible switching of emulsification/breaking behavior could be reached at least 10 times. This "intelligent emulsifier" exhibited high emulsification efficiency, long-term stability, and on-demand emulsification/breaking properties. It was notable that MNPDNML microspheres showed excellent emulsification ability for olive oil, kerosene, gasoline, and crude oil, which allowed the material to be widely used in the controlled transportation and separation of fuel oil.

4.
Theranostics ; 14(5): 2190-2209, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38505600

RESUMO

Here we explored the potential role of Gαi2 (G protein subunit alpha i2) in endothelial cell function and angiogenesis. Methods: Genetic methodologies such as shRNA, CRISPR/Cas9, dominant negative mutation, and overexpression were utilized to modify Gαi2 expression or regulate its function. Their effects on endothelial cell functions were assessed in vitro. In vivo, the endothelial-specific Gαi2 shRNA adeno-associated virus (AAV) was utilized to silence Gαi2 expression. The impact of this suppression on retinal angiogenesis in control mice and streptozotocin (STZ)-induced diabetic retinopathy (DR) mice was analyzed. Results: Analysis of single-cell RNA sequencing data revealed Gαi2 (GNAI2) was predominantly expressed in retinal endothelial cells and expression was increased in retinal endothelial cells following oxygen-induced retinopathy (OIR) in mice. Moreover, transcriptome analysis linking Gαi2 to angiogenesis-related processes/pathways, supported by increased Gαi2 expression in experimental OIR mouse retinas, highlighted its possible role in angiogenesis. In various endothelial cell types, shRNA-induced silencing and CRISPR/Cas9-mediated knockout (KO) of Gαi2 resulted in substantial reductions in cell proliferation, migration, invasion, and capillary tube formation. Conversely, Gαi2 over-expression in endothelial cells induced pro-angiogenic activities, enhancing cell proliferation, migration, invasion, and capillary tube formation. Furthermore, our investigation revealed a crucial role of Gαi2 in NFAT (nuclear factor of activated T cells) activation, as evidenced by the down-regulation of NFAT-luciferase reporter activity and pro-angiogenesis NFAT-targeted genes (Egr3, CXCR7, and RND1) in Gαi2-silenced or -KO HUVECs, which were up-regulated in Gαi2-overexpressing endothelial cells. Expression of a dominant negative Gαi2 mutation (S48C) also down-regulated NFAT-targeted genes, slowing proliferation, migration, invasion, and capillary tube formation in HUVECs. Importantly, in vivo experiments revealed that endothelial Gαi2 knockdown inhibited retinal angiogenesis in mice, with a concomitant down-regulation of NFAT-targeted genes in mouse retinal tissue. In contrast, Gαi2 over-expression in endothelial cells enhanced retinal angiogenesis in mice. Single-cell RNA sequencing data confirmed increased levels of Gαi2 specifically in retinal endothelial cells of mice with streptozotocin (STZ)-induced diabetic retinopathy (DR). Importantly, endothelial Gαi2 silencing ameliorated retinal pathological angiogenesis in DR mice. Conclusion: Our study highlights a critical role for Gαi2 in NFAT activation, endothelial cell activation and angiogenesis, offering valuable insights into potential therapeutic strategies for modulating these processes.


Assuntos
Retinopatia Diabética , Camundongos , Animais , Retinopatia Diabética/tratamento farmacológico , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/metabolismo , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/farmacologia , Células Endoteliais/metabolismo , Angiogênese , Estreptozocina/efeitos adversos , Oxigênio/metabolismo , RNA Interferente Pequeno/metabolismo , Proliferação de Células
5.
Front Endocrinol (Lausanne) ; 14: 1276225, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37842315

RESUMO

Macrophages/microglia are immune system defense and homeostatic cells that develop from bone marrow progenitor cells. According to the different phenotypes and immune responses of macrophages (Th1 and Th2), the two primary categories of polarized macrophages/microglia are those conventionally activated (M1) and alternatively activated (M2). Macrophage/microglial polarization is a key regulating factor in the development of inflammatory disorders, cancers, metabolic disturbances, and neural degeneration. Macrophage/microglial polarization is involved in inflammation, oxidative stress, pathological angiogenesis, and tissue healing processes in ocular diseases, particularly in diabetic retinopathy (DR). The functional phenotypes of macrophages/microglia affect disease progression and prognosis, and thus regulate the polarization or functional phenotype of microglia at different DR stages, which may offer new concepts for individualized therapy of DR. This review summarizes the involvement of macrophage/microglia polarization in physiological situations and in the pathological process of DR, and discusses the promising role of polarization in personalized treatment of DR.


Assuntos
Diabetes Mellitus , Retinopatia Diabética , Humanos , Microglia/metabolismo , Retinopatia Diabética/metabolismo , Macrófagos/metabolismo , Inflamação/metabolismo , Ativação de Macrófagos , Diabetes Mellitus/metabolismo
6.
Cell Death Dis ; 14(10): 700, 2023 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-37880221

RESUMO

We here tested the potential activity and the underlying mechanisms of neuroligin-3 (NLGN3) against ischemia-reperfusion-induced neuronal cell injury. In SH-SY5Y neuronal cells and primary murine cortical neurons, NLGN3 activated Akt-mTOR and Erk signalings, and inhibited oxygen and glucose deprivation (OGD)/re-oxygenation (OGD/R)-induced cytotoxicity. Akt activation was required for NLGN3-induced neuroprotection. Gαi1/3 mediated NLGN3-induced downstream signaling activation. NLGN3-induced Akt-S6K1 activation was largely inhibited by Gαi1/3 silencing or knockout. Significantly, NLGN3-induced neuroprotection against OGD/R was almost abolished by Gαi1/3 silencing or knockout. In vivo, the middle cerebral artery occlusion (MCAO) procedure induced NLGN3 cleavage and secretion, and increased its expression and Akt activation in mouse brain tissues. ADAM10 (A Disintegrin and Metalloproteinase 10) inhibition blocked MCAO-induced NLGN3 cleavage and secretion, exacerbating ischemic brain injury in mice. Neuronal silencing of NLGN3 or Gαi1/3 in mice also inhibited Akt activation and intensified MCAO-induced ischemic brain injury. Conversely, neuronal overexpression of NLGN3 increased Akt activation and alleviated MCAO-induced ischemic brain injury. Together, NLGN3 activates Gαi1/3-Akt signaling to protect neuronal cells from ischemia-reperfusion injury.


Assuntos
Lesões Encefálicas , Isquemia Encefálica , Neuroblastoma , Traumatismo por Reperfusão , Animais , Humanos , Camundongos , Lesões Encefálicas/metabolismo , Isquemia Encefálica/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Neuroblastoma/metabolismo , Neurônios/metabolismo , Oxigênio/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Traumatismo por Reperfusão/metabolismo
7.
Theranostics ; 13(7): 2319-2336, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37153740

RESUMO

Netrin-1 binds to the high-affinity receptor CD146 to activate downstream signaling and angiogenesis. Here, we examine the role and underlying mechanisms of G protein subunit alpha i1 (Gαi1) and Gαi3 in Netrin-1-induced signaling and pro-angiogenic activity. In mouse embryonic fibroblasts (MEFs) and endothelial cells, Netrin-1-induced Akt-mTOR (mammalian target of rapamycin) and Erk activation was largely inhibited by silencing or knockout of Gαi1/3, whereas signaling was augmented following Gαi1/3 overexpression. Netrin-1 induced Gαi1/3 association with CD146, required for CD146 internalization, Gab1 (Grb2 associated binding protein 1) recruitment and downstream Akt-mTOR and Erk activation. Netrin-1-induced signaling was inhibited by CD146 silencing, Gab1 knockout, or Gαi1/3 dominant negative mutants. Netrin-1-induced human umbilical vein endothelial cell (HUVEC) proliferation, migration and tube formation were inhibited by Gαi1/3 short hairpin RNA (shRNA), but were potentiated by ectopic Gαi1/3 overexpression. In vivo, intravitreous injection of Netrin-1 shRNA adeno-associated virus (AAV) significantly inhibited Akt-mTOR and Erk activation in murine retinal tissues and reduced retinal angiogenesis. Endothelial knockdown of Gαi1/3 significantly inhibited Netrin1-induced signaling and retinal angiogenesis in mice. Netrin-1 mRNA and protein expression were significantly elevated in retinal tissues of diabetic retinopathy (DR) mice. Importantly, silence of Netrin-1, by intravitreous Netrin-1 shRNA AAV injection, inhibited Akt-Erk activation, pathological retinal angiogenesis and retinal ganglion cells degeneration in DR mice. Lastly, Netrin-1 and CD146 expression is significantly increased in the proliferative retinal tissues of human proliferative diabetic retinopathy patients. Together, Netrin-1 induces CD146-Gαi1/3-Gab1 complex formation to mediate downstream Akt-mTOR and Erk activation, important for angiogenesis in vitro and in vivo.


Assuntos
Retinopatia Diabética , Proteínas Proto-Oncogênicas c-akt , Humanos , Animais , Camundongos , Antígeno CD146/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Netrina-1 , Fibroblastos/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , RNA Interferente Pequeno , Proteínas de Transporte , Mamíferos/metabolismo
8.
Int J Biol Sci ; 19(6): 1910-1924, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37063428

RESUMO

The stem cell factor (SCF) binds to c-Kit in endothelial cells, thus activating downstream signaling and angiogenesis. Herein, we examined the role of G protein subunit alpha inhibitory (Gαi) proteins in this process. In MEFs and HUVECs, Gαi1/3 was associated with SCF-activated c-Kit, promoting c-Kit endocytosis, and binding of key adaptor proteins, subsequently transducing downstream signaling. SCF-induced Akt-mTOR and Erk activation was robustly attenuated by Gαi1/3 silencing or knockout (KO), or due to dominant negative mutations but was strengthened substantially following ectopic overexpression of Gαi1/3. SCF-induced HUVEC proliferation, migration, and capillary tube formation were suppressed after Gαi1/3 silencing or KO, or due to dominant negative mutations. In vivo, endothelial knockdown of Gαi1/3 by intravitreous injection of endothelial-specific shRNA adeno-associated virus (AAV) potently reduced SCF-induced signaling and retinal angiogenesis in mice. Moreover, mRNA and protein expressions of SCF increased significantly in the retinal tissues of streptozotocin-induced diabetic retinopathy (DR) mice. SCF silencing, through intravitreous injection of SCF shRNA AAV, inhibited pathological retinal angiogenesis and degeneration of retinal ganglion cells in DR mice. Finally, the expression of SCF and c-Kit increased in proliferative retinal tissues of human patients with proliferative DR. Taken together, Gαi1/3 mediate SCF/c-Kit-activated signaling and angiogenesis.


Assuntos
Células Endoteliais , Transdução de Sinais , Animais , Humanos , Camundongos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Células Endoteliais/metabolismo , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/genética , Fator de Células-Tronco/genética , Fator de Células-Tronco/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo
10.
J Oncol ; 2022: 8660965, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36262353

RESUMO

Background: Osteosarcoma (OS) is a malignant tumor that is highly metastatic with a high mortality rate. Although mounting evidence suggests that LINC00909 is strongly associated with the malignant progression of various tumors, the exact role of LINC00909 in OS remains unknown. Therefore, the current study was designed to investigate the relationship between LINC00909 and the malignant progression of OS. Methods: LINC00909 expression was measured in OS cell lines and clinical specimens using RT-qPCR assays. The effects of LINC00909 on OS proliferation, invasion, and migration were calculated both in vitro and in vivo. Apart from that, bioinformatics analyses, FISH, RIP, and luciferase reporter assays were carried out to investigate the downstream target of LINC00909. Rescue experiments were also conducted to investigate the potential mechanisms of action of competitive endogenous RNAs (ceRNAs). Results: In this study, we found that LINC00909 was highly expressed in OS cell lines and clinical specimens. In vivo and in vitro experiments demonstrated that LINC00909 induces epithelial-to-mesenchymal transition (EMT) and contributes to OS tumorigenesis and metastasis. FISH, RIP, and luciferase assays indicated that miR-875-5p is a direct target of LINC00909. Moreover, HOXD9 was validated as the downstream target of miR-875-5p in a luciferase reporter assay and western blotting experiments. Rescue experiments revealed that HOXD9 reversed the effect of LV-sh-LINC00909 on OS cells by positively regulating the PI3K/AKT/mTOR signaling pathway. Conclusion: Collectively, LINC00909 induces EMT and contributes to OS tumorigenesis and metastasis through the PI3K/AKT/mTOR pathway by binding to miR-875-5p to upregulate HOXD9 expression. Targeting the LINC00909/miR-875-5p/HOXD9 axis may have potential in treating OS.

11.
Theranostics ; 11(17): 8535-8549, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34373757

RESUMO

Neuroligin-3 (NLGN3) is necessary and sufficient to promote glioma cell growth. The recruitment of Gαi1/3 to the ligand-activated receptor tyrosine kinases (RTKs) is essential for mediating oncogenic signaling. Methods: Various genetic strategies were utilized to examine the requirement of Gαi1/3 in NLGN3-driven glioma cell growth. Results: NLGN3-induced Akt-mTORC1 and Erk activation was inhibited by decreasing Gαi1/3 expression. In contrast ectopic Gαi1/3 overexpression enhanced NLGN3-induced signaling. In glioma cells, NLGN3-induced cell growth, proliferation and migration were attenuated by Gαi1/3 depletion with shRNA, but facilitated with Gαi1/3 overexpression. Significantly, Gαi1/3 silencing inhibited orthotopic growth of patient-derived glioma xenografts in mouse brain, whereas forced Gαi1/3-overexpression in primary glioma xenografts significantly enhanced growth. The growth of brain-metastatic human lung cancer cells in mouse brain was largely inhibited with Gαi1/3 silencing. It was however expedited with ectopic Gαi1/3 overexpression. In human glioma Gαi3 upregulation was detected, correlating with poor prognosis. Conclusion: Gαi1/3 mediation of NLGN3-induced signaling is essential for neuronal-driven glioma growth.


Assuntos
Moléculas de Adesão Celular Neuronais/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Glioma/metabolismo , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Idoso , Animais , Neoplasias Encefálicas/patologia , Moléculas de Adesão Celular Neuronais/fisiologia , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/fisiologia , Glioma/genética , Glioma/patologia , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Proteínas de Membrana/fisiologia , Camundongos , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/fisiologia , Neurônios/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Extratos Vegetais , Cultura Primária de Células , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais
12.
Theranostics ; 11(10): 4894-4909, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33754034

RESUMO

IL-4 induces Akt activation in macrophages, required for full M2 (alternative) polarization. We examined the roles of Gαi1 and Gαi3 in M2 polarization using multiple genetic methods. Methods and Results: In MEFs and primary murine BMDMs, Gαi1/3 shRNA, knockout or dominant negative mutations attenuated IL-4-induced IL4Rα endocytosis, Gab1 recruitment as well as Akt activation, leaving STAT6 signaling unaffected. Following IL-4 stimulation, Gαi1/3 proteins associated with the intracellular domain of IL-4Rα and the APPL1 adaptor, to mediate IL-4Rα endosomal traffic and Gab1-Akt activation in BMDMs. In contrast, gene silencing of Gαi1/3 with shRNA or knockout resulted in BMDMs that were refractory to IL-4-induced M2 polarization. Conversely, Gαi1/3-overexpressed BMDMs displayed preferred M2 response with IL-4 stimulation. In primary human macrophages IL-4-induced Akt activation and Th2 genes expression were inhibited with Gαi1/3 silencing, but augmented with Gαi1/3 overexpression. In Gαi1/3 double knockout (DKO) mice, M2 polarization, by injection of IL-4 complex or chitin, was potently inhibited. Moreover, in a murine model of asthma, ovalbumin-induced airway inflammation and hyperresponsiveness were largely impaired in Gαi1/3 DKO mice. Conclusion: These findings highlight novel and essential roles for Gαi1/3 in regulating IL-4-induced signaling, macrophage M2 polarization and allergic asthma response.


Assuntos
Asma/imunologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Interleucina-4/imunologia , Macrófagos/imunologia , Hipersensibilidade Respiratória/genética , Animais , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/imunologia , Camundongos , Camundongos Knockout , Ovalbumina , Proteínas Proto-Oncogênicas c-akt/metabolismo , Hipersensibilidade Respiratória/imunologia , Serina-Treonina Quinases TOR/metabolismo
13.
Dis Markers ; 2021: 6575195, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34987675

RESUMO

BACKGROUND: The vitreous body is an important part of the ocular body fluid. A foldable capsular vitreous body (FCVB) is designed to treat chronic adverse complications in severe ocular trauma and silicone oil-dependent eyes. This study is aimed at investigating a method for implanting an FCVB, its postoperative efficacy, and clinical value. METHODS: A retrospective analysis was performed on data from 18 patients who underwent vitrectomy and FCVB implantation for severe ocular trauma and silicone oil-dependent eyes between March 2019 and May 2020. All treated eyes underwent clinical examinations involving the best-corrected visual acuity, intraocular pressure, FCVB position, anterior segment photography, and wide-angle fundus photography regularly after surgery. RESULTS: Eighteen eyes from 18 patients were enrolled in this study. A total of 2.00-4.20 (3.46 ± 0.78) ml of silicone oil were injected into the FCVB during surgery. The patients were followed up at 1, 2, and 4 weeks and 3, 6, and 12 months after surgery. Twelve months after surgery, visual acuity improved in 7 (38.89%) eyes. In contrast, 10 (55.56%) eyes showed no obvious improvement, and 1 (5.56%) eye had decreased vision. Intraocular pressure at 12 months was 10.13 ± 3.52 mmHg, which was comparable to that before the surgery (t = 0.38, P = 0.71). The anterior chamber depth examined by slit lamp was 2.00-3.00 cornea thickness (CT) in 7 eyes, 1.00-2.00 CT in 2 eyes, and <1.00 CT in one eye. The anterior chamber disappeared in eight eyes. There were eight eyes with clear cornea, four eyes with localized opacity, and two eyes with obvious gray-white opacity. There was no case of severe FCVB deflection, rupture, or exposure during the observation period. CONCLUSION: FCVB implantation is an effective and safe treatment for eyes with severe ocular trauma and silicone oil-dependent eyes. It may support retinal reattachment, slow down eyeball atrophy, reduce the risk of chronic adverse complications such as corneal endothelial decompensation, and maintain intraocular pressure and preoperative visual function.


Assuntos
Óleos de Silicone , Vitrectomia , Corpo Vítreo/transplante , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Resultado do Tratamento
14.
Biochem Biophys Res Commun ; 514(3): 777-784, 2019 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-31079921

RESUMO

Ultra-violet radiation (UVR) can induce significant oxidative injury to human lens epithelial cells (HLECs). Sirtuin 6 (SIRT6) is shown to directly bind to Nrf2, essential for Nrf2 signaling activation. In the present study, we show that microRNA-4532 (miR-4532) targets SIRT6 to regulate Nrf2 signaling in HLECs. Ectopic overexpression of miR-4532 in HLECs decreased SIRT6 3'-UTR activity, causing SIRT6 downregulation and Nrf2 signaling inhibition. Conversely, miR-4532 inhibition, by a lentiviral construct, enhanced SIRT6 3'-UTR activity, SIRT6 expression and Nrf2 signaling activation. Functional studies show that UVR-induced cytotoxicity and apoptosis in HLECs were potentiated by miR-4532 overexpression, Nrf2 depletion or SIRT6 shRNA. Conversely, miR-4532 inhibition or ectopic SIRT6 overexpression attenuated UVR-induced oxidative injury in HLECs. Importantly, miR-4532 overexpression or inhibition was ineffective in SIRT6-KO or Nrf2-KO HLECs. Taken together, the results show that inhibition of miR-4532 protects HLECs from UVR-induced oxidative injury via activation of SIRT6-Nrf2 pathway. Targeting the miR-4532-SIRT6-Nrf2 pathway could be a novel strategy to protect HLECs from UVR and possible other oxidative stresses.


Assuntos
Citoproteção , Células Epiteliais/patologia , MicroRNAs/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo/efeitos da radiação , Transdução de Sinais , Sirtuínas/metabolismo , Raios Ultravioleta , Sequência de Bases , Citoproteção/efeitos da radiação , Células Epiteliais/metabolismo , Células Epiteliais/efeitos da radiação , Humanos , MicroRNAs/genética , Transdução de Sinais/efeitos da radiação
15.
Theranostics ; 8(17): 4695-4709, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30279732

RESUMO

VEGF binding to VEGFR2 leads to VEGFR2 endocytosis and downstream signaling activation to promote angiogenesis. Methods: Using genetic strategies, we tested the requirement of α subunits of heterotrimeric G proteins (Gαi1/3) in the process. Results: Gαi1/3 are located in the VEGFR2 endocytosis complex (VEGFR2-Ephrin-B2-Dab2-PAR-3), where they are required for VEGFR2 endocytosis and downstream signaling transduction. Gαi1/3 knockdown, knockout or dominant negative mutation inhibited VEGF-induced VEGFR2 endocytosis, and downstream Akt-mTOR and Erk-MAPK activation. Functional studies show that Gαi1/3 shRNA inhibited VEGF-induced proliferation, invasion, migration and vessel-like tube formation of HUVECs. In vivo, Gαi1/3 shRNA lentivirus inhibited alkali burn-induced neovascularization in mouse cornea. Further, oxygen-induced retinopathy (OIR)-induced retinal neovascularization was inhibited by intravitreal injection of Gαi1/3 shRNA lentivirus. Moreover, in vivo angiogenesis by alkali burn and OIR was significantly attenuated in Gαi1/3 double knockout mice. Significantly, Gαi1/3 proteins are upregulated in proliferative retinal tissues of proliferative diabetic retinopathy (PDR) patients. Conclusion: These results provide mechanistic insights into the critical role played by Gαi1/3 proteins in VEGF-induced VEGFR2 endocytosis, signaling and angiogenesis.


Assuntos
Endocitose , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Neovascularização Fisiológica , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Técnicas de Silenciamento de Genes , Técnicas de Inativação de Genes , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Modelos Animais , Modelos Teóricos , Ligação Proteica
16.
FASEB J ; 32(7): 3782-3791, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29465315

RESUMO

VEGF-induced neovascularization plays a pivotal role in corneal neovascularization (CoNV). The current study investigated the potential effect of ginsenoside Rh2 (GRh2) on neovascularization. In HUVECs, pretreatment with GRh2 largely attenuated VEGF-induced cell proliferation, migration, and vessel-like tube formation in vitro. At the molecular level, GRh2 disrupted VEGF-induced VEGF receptor 2 (VEGFR2)-Grb-2-associated binder 1 (Gab1) association in HUVECs, causing inactivation of downstream AKT and ERK signaling. Gab1 knockdown (by targeted short hairpin RNA) similarly inhibited HUVEC proliferation and migration. Notably, GRh2 was ineffective against VEGF in Gab1-silenced HUVECs. In a mouse cornea alkali burn model, GRh2 eyedrops inhibited alkali-induced neovascularization and inflammatory cell infiltrations in the cornea. Furthermore, alkali-induced corneal expression of mRNAs/long noncoding RNAs in cornea were largely attenuated by GRh2. Overall, GRh2 inhibits VEGF-induced angiogenic effect via inhibiting VEGFR2-Gab1 signaling in vitro. It also alleviates angiogenic and inflammatory responses in alkali burn-treated mouse corneas.-Zhang, X.-P., Li, K.-R., Yu, Q., Yao, M.-D., Ge, H.-M., Li, X.-M., Jiang, Q., Yao, J., Cao, C. Ginsenoside Rh2 inhibits vascular endothelial growth factor-induced corneal neovascularization.


Assuntos
Anti-Inflamatórios/farmacologia , Neovascularização da Córnea/tratamento farmacológico , Ginsenosídeos/farmacologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Anti-Inflamatórios/uso terapêutico , Córnea/efeitos dos fármacos , Córnea/metabolismo , Neovascularização da Córnea/etiologia , Neovascularização da Córnea/metabolismo , Ginsenosídeos/uso terapêutico , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Gelo , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos Endogâmicos ICR , Fosfoproteínas/metabolismo , Fator A de Crescimento do Endotélio Vascular/toxicidade , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
17.
Free Radic Biol Med ; 117: 238-246, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29427790

RESUMO

Excessive Ultra-violet (UV) radiation shall induce damages to resident retinal pigment epithelium (RPE) cells (RPEs) and retinal ganglion cells (RGCs). Here we tested the potential activity of Ginsenoside Rh3 ("Rh3") against the process. In cultured human RPEs and RGCs, pretreatment with Rh3 inhibited UV-induced reactive oxygen species (ROS) production and following apoptotic/non-apoptotic cell death. Rh3 treatment in retinal cells induced nuclear-factor-E2-related factor 2 (Nrf2) activation, which was evidenced by Nrf2 protein stabilization and its nuclear translocation, along with transcription of antioxidant responsive element (ARE)-dependent genes (HO1, NOQ1 and GCLC). Nrf2 knockdown by targeted-shRNA almost abolished Rh3-induced retinal cell protection against UV. Further studies found that Rh3 induced microRNA-141 ("miR-141") expression, causing downregulation of its targeted gene Keap1 in RPEs and RGCs. On the other hand, Rh3-induced Nrf2 activation and retinal cell protection were largely attenuated by the miR-141's inhibitor, antagomiR-141. In vivo, intravitreal injection of Rh3 inhibited retinal dysfunction by light damage in mice. Rh3 intravitreal injection also induced miR-141 expression, Keap1 downregulation and Nrf2 activation in mouse retinas. We conclude that Rh3 protects retinal cells from UV via activating Nrf2 signaling.


Assuntos
Ginsenosídeos/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo , Células Ganglionares da Retina/efeitos dos fármacos , Epitélio Pigmentado da Retina/efeitos dos fármacos , Raios Ultravioleta/efeitos adversos , Animais , Ativação Enzimática/efeitos dos fármacos , Humanos , Camundongos , Fator 2 Relacionado a NF-E2/efeitos dos fármacos , Lesões Experimentais por Radiação/tratamento farmacológico
18.
Oncotarget ; 7(37): 60123-60132, 2016 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-27517753

RESUMO

Excessive Ultra-violet (UV) radiation causes oxidative damages and apoptosis in retinal pigment epithelium (RPE) cells. Here we tested the potential activity of SC79, a novel small molecule activator of Akt, against the process. We showed that SC79 activated Akt in primary and established (ARPE-19 line) RPE cells. It protected RPE cells from UV damages possibly via inhibiting cell apoptosis. Akt inhibition, via an Akt specific inhibitor (MK-2206) or Akt1 shRNA silence, almost abolished SC79-induced RPE cytoprotection. Further studies showed that SC79 activated Akt-dependent NF-E2-related factor 2 (Nrf2) signaling and inhibited UV-induced oxidative stress in RPE cells. Reversely, Nrf2 shRNA knockdown or S40T mutation attenuated SC79-induced anti-UV activity. For the in vivo studies, we showed that intravitreal injection of SC79 significantly protected mouse retina from light damages. Based on these results, we suggest that SC79 protects RPE cells from UV damages possibly via activating Akt-Nrf2 signaling axis.


Assuntos
Acetatos/farmacologia , Benzopiranos/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Epitélio Pigmentado da Retina/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Acetatos/química , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Apoptose/efeitos da radiação , Benzopiranos/química , Linhagem Celular , Células Cultivadas , Compostos Heterocíclicos com 3 Anéis/farmacologia , Humanos , Masculino , Camundongos Endogâmicos BALB C , Estrutura Molecular , Mutação , Fator 2 Relacionado a NF-E2/genética , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/efeitos da radiação , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/genética , Interferência de RNA , Epitélio Pigmentado da Retina/citologia , Epitélio Pigmentado da Retina/efeitos da radiação , Transdução de Sinais/genética , Transdução de Sinais/efeitos da radiação , Raios Ultravioleta
19.
Sci Rep ; 6: 25525, 2016 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-27151674

RESUMO

Excessive UV radiation and reactive oxygen species (ROS) cause retinal pigment epithelium (RPE) cell injuries. Nrf2 regulates transcriptional activation of many anti-oxidant genes. Here, we tested the potential role of 3H-1,2-dithiole-3-thione (D3T) against UV or ROS damages in cultured RPE cells (both primary cells and ARPE-19 line). We showed that D3T significantly inhibited UV-/H2O2-induced RPE cell death and apoptosis. UV-stimulated ROS production was dramatically inhibited by D3T pretreatment. D3T induced Nrf2 phosphorylation in cultured RPE cells, causing Nrf2 disassociation with KEAP1 and its subsequent nuclear accumulation. This led to expression of antioxidant response elements (ARE)-dependent gene heme oxygenase-1 (HO-1). Nrf2-HO-1 activation was required for D3T-mediated cytoprotective effect. Nrf2 shRNA knockdown or S40T dominant negative mutation as well as the HO-1 inhibitor Zinc protoporphyrin (ZnPP) largely inhibited D3T's RPE cytoprotective effects against UV radiation. Yet, exogenous overexpression Nrf2 enhanced D3T's activity in RPE cells. Further studies showed that D3T activated Akt/mTORC1 in cultured RPE cells. Akt-mTORC1 inhibitors, or Akt1 knockdown by shRNA, not only inhibited D3T-induced Nrf2-HO-1 activation, but also abolished the RPE cytoprotective effects. In vivo, D3T intravitreal injection protected from light-induced retinal dysfunctions in mice. Thus, D3T protects RPE cells from UV-induced damages via activation of Akt-mTORC1-Nrf2-HO-1 signaling axis.


Assuntos
Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Protetores contra Radiação/metabolismo , Epitélio Pigmentado da Retina/efeitos da radiação , Tionas/metabolismo , Tiofenos/metabolismo , Raios Ultravioleta , Animais , Apoptose , Células Cultivadas , Heme Oxigenase-1 , Humanos , Proteínas de Membrana , Camundongos Endogâmicos C57BL , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
20.
Am J Physiol Endocrinol Metab ; 307(12): E1153-65, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25352436

RESUMO

In response to hyperglycemia in patients with diabetes, many signaling pathways contribute to the pathogenesis of diabetic complications, including diabetic retinopathy (DR). Excessive production of inflammatory mediators plays an important role in this process. Amadori-glycated albumin, one of the major forms of advanced glycated end-products, has been implicated in DR by inducing inflammatory responses in microglia/macrophages. Our goal was to delineate the potential cross talk between class A scavenger receptor (SR-A) and the receptor for advanced glycated end-product (RAGE) in the context of DR. We show here that SR-A ablation caused an exacerbated form of DR in streptozotocin-injected C57BL/6J mice as evidenced by fundus imaging and electroretinography. Immunohistochemical staining and RT-PCR assay indicated that there was augmented activation of proinflammatory macrophages with upregulated synthesis of proinflammatory mediators in the retina in Sr-a(-/-) mice. Overexpression of SR-A suppressed RAGE-induced mitogen-activated protein kinase (MAPK) signaling, whereas RAGE activation in macrophages favored a proinflammatory (M1) phenotype in the absence of SR-A. Mechanistic analysis on bone marrow-derived macrophages and HEK293 cell line revealed that SR-A interacted with and inhibited the phosphorylation of mitogen-activated protein kinase kinase 7, the major kinase in the RAGE-MAPK-NF-κB signaling, thereby leading to diminished secretion of proinflammatory cytokines. Our findings suggest that the antagonism between SR-A and RAGE contributes to the pathogenesis of DR by nurturing a disease-prone macrophage phenotype. Therefore, specific agonist that boosts SR-A signaling could potentially provide benefits in the prevention and/or intervention of DR.


Assuntos
Retinopatia Diabética/genética , Receptor Cross-Talk/fisiologia , Receptores Imunológicos/fisiologia , Receptores Depuradores Classe A/fisiologia , Animais , Células Cultivadas , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Retinopatia Diabética/patologia , Células HEK293 , Humanos , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor para Produtos Finais de Glicação Avançada , Transdução de Sinais/genética , Estreptozocina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA