Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.301
Filtrar
1.
Cell Mol Biol Lett ; 29(1): 47, 2024 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-38589823

RESUMO

BACKGROUND: Glucose-6-phosphate dehydrogenase (G6PD) plays an important role in vascular smooth muscle cell (VSMC) phenotypic switching, which is an early pathogenic event in various vascular remodeling diseases (VRDs). However, the underlying mechanism is not fully understood. METHODS: An IP‒LC‒MS/MS assay was conducted to identify new binding partners of G6PD involved in the regulation of VSMC phenotypic switching under platelet-derived growth factor-BB (PDGF-BB) stimulation. Co-IP, GST pull-down, and immunofluorescence colocalization were employed to clarify the interaction between G6PD and voltage-dependent anion-selective channel protein 1 (VDAC1). The molecular mechanisms involved were elucidated by examining the interaction between VDAC1 and apoptosis-related biomarkers, as well as the oligomerization state of VDAC1. RESULTS: The G6PD level was significantly elevated and positively correlated with the synthetic characteristics of VSMCs induced by PDGF-BB. We identified VDAC1 as a novel G6PD-interacting molecule essential for apoptosis. Specifically, the G6PD-NTD region was found to predominantly contribute to this interaction. G6PD promotes VSMC survival and accelerates vascular neointimal hyperplasia by inhibiting VSMC apoptosis. Mechanistically, G6PD interacts with VDAC1 upon stimulation with PDGF-BB. By competing with Bax for VDAC1 binding, G6PD reduces VDAC1 oligomerization and counteracts VDAC1-Bax-mediated apoptosis, thereby accelerating neointimal hyperplasia. CONCLUSION: Our study showed that the G6PD-VDAC1-Bax axis is a vital switch in VSMC apoptosis and is essential for VSMC phenotypic switching and neointimal hyperplasia, providing mechanistic insight into early VRDs.


Assuntos
Glucosefosfato Desidrogenase , Músculo Liso Vascular , Canal de Ânion 1 Dependente de Voltagem , Humanos , Hiperplasia/metabolismo , Hiperplasia/patologia , Becaplermina/genética , Becaplermina/metabolismo , Proliferação de Células , Proteína X Associada a bcl-2/metabolismo , Glucosefosfato Desidrogenase/metabolismo , Músculo Liso Vascular/metabolismo , Cromatografia Líquida , Espectrometria de Massas em Tandem , Neointima/genética , Neointima/metabolismo , Neointima/patologia , Apoptose , Miócitos de Músculo Liso/metabolismo , Movimento Celular/genética , Células Cultivadas , Fenótipo
2.
Sci Rep ; 14(1): 7947, 2024 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-38575707

RESUMO

Graves' ophthalmopathy (GO) is an extra-thyroidal complication of Graves' disease which can lead to vision loss in severe cases. Currently, treatments of GO are not sufficiently effective, so novel therapeutic strategies are needed. As platelet-derived growth factor (PDGF)-BB induces several effector mechanisms in GO orbital fibroblasts including cytokine production and myofibroblast activation, this study aims to investigate the roles of histone lysine methyltransferases (HKMTs) in PDGF-BB-activated GO orbital fibroblasts by screening with HKMTs inhibitors library. From the total of twelve selective HKMT inhibitors in the library, EZH2, G9a and DOT1L inhibitors, DZNeP, BIX01294 and Pinometostat, respectively, prevented PDGF-BB-induced proliferation and hyaluronan production by GO orbital fibroblasts. However, only EZH2 inhibitor, DZNeP, significantly blocked pro-inflammatory cytokine production. For the HKMTs expression in GO orbital fibroblasts, PDGF-BB significantly and time-dependently induced EZH2, G9a and DOT1L mRNA expression. To confirm the role of EZH2 in PDGF-BB-induced orbital fibroblast activation, EZH2 silencing experiments revealed suppression of PDGF-BB-induced collagen type I and α-SMA expression along with decreasing histone H3 lysine 27 trimethylation (H3K27me3) level. In a more clinically relevant model than orbital fibroblast culture experiments, DZNeP treated GO orbital tissues significantly reduced pro-inflammatory cytokine production while slightly reduced ACTA2 mRNA expression. Our data is the first to demonstrate that among all HKMTs EZH2 dominantly involved in the expression of myofibroblast markers in PDGF-BB-activated orbital fibroblast from GO presumably via H3K27me3. Thus, EZH2 may represent a novel therapeutics target for GO.


Assuntos
Oftalmopatia de Graves , Histonas , Humanos , Becaplermina/metabolismo , Proteínas Proto-Oncogênicas c-sis/genética , Histona Metiltransferases/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Órbita/patologia , Oftalmopatia de Graves/metabolismo , Citocinas/metabolismo , Fibroblastos/metabolismo , RNA Mensageiro/genética , Células Cultivadas , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo
3.
Int J Mol Sci ; 25(7)2024 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-38612456

RESUMO

Platelets are actively involved in tissue injury site regeneration by producing a wide spectrum of platelet-derived growth factors such as PDGF (platelet-derived growth factor), IGF-1 (insulin-like growth factor), TGF-ß1 (transforming growth factor ß), FGF (fibroblast growth factor), etc. A rotating magnetic field (RMF) can regulate biological functions, including reduction or induction regarding inflammatory processes, cell differentiation, and gene expression, to determine the effect of an RMF on the regenerative potential of platelets. The study group consisted of 30 healthy female and male volunteers (n = 15), from which plasma was collected. A portion of the plasma was extracted and treated as an internal control group. Subsequent doses of plasma were exposed to RMF at different frequencies (25 and 50 Hz) for 1 and 3 h. Then, the concentrations of growth factors (IGF-1, PDGF-BB, TGF-ß1, and FGF-1) were determined in the obtained material by the ELISA method. There were statistically significant differences in the PDGF-BB, TGF-ß1, IGF-1, and FGF-1 concentrations between the analyzed groups. The highest concentration of PDGF-BB was observed in the samples placed in RMF for 1 h at 25 Hz. For TGF-ß1, the highest concentrations were obtained in the samples exposed to RMF for 3 h at 25 Hz and 1 h at 50 Hz. The highest concentrations of IGF-1 and FGF-1 were shown in plasma placed in RMF for 3 h at 25 Hz. An RMF may increase the regenerative potential of platelets. It was noted that female platelets may respond more strongly to RMF than male platelets.


Assuntos
Fator 1 de Crescimento de Fibroblastos , Fator de Crescimento Insulin-Like I , Humanos , Feminino , Masculino , Becaplermina , Fator de Crescimento Transformador beta1 , Fatores de Crescimento de Fibroblastos , Fator de Crescimento Derivado de Plaquetas , Campos Magnéticos
4.
FASEB J ; 38(6): e23557, 2024 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-38498343

RESUMO

Phenotypic switching of vascular smooth muscle cells (VSMCs) is essential for atherosclerosis development. Circular RNA (circRNA) is a specific non-coding RNA that is produced as a closed-loop structure in mammals, and its specific expression pattern is closely related to its cell type and tissue. To clarify the roles of circTLK1 in VSMC phenotypic switching, we performed qRT-PCR, immunoblotting, and immunostaining. qRT-PCR revealed that circTLK1 was upregulated in both mouse models of atherosclerosis in vivo and PDGF (platelet-derived growth factor)-BB-induced VSMCs in vitro. Furthermore, the overexpression of circTLK1 promoted PDGF-BB-induced VSMC phenotypic switching. Conversely, experiments performed in vivo demonstrate that the knockdown of SMC-specific circTLK1 led to a reduction in the development of atherosclerosis. The relationship between circTLK1 and miR-513a-3p and Krüppel-like factor 4 (KLF4) was detected by RNA immunoprecipitation (RIP), luciferase reporter assay, RNA pull-down, and RNA fluorescence in situ hybridization (RNA FISH). Mechanistically, circTLK1 acted as a sponge for miR-513a-3p, leading to the upregulation of KLF4, a key transcription factor for phenotypic switching. Targeting the circTLK1/miR-513a-3p/KLF4 axis may provide a potential therapeutic strategy for atherosclerosis.


Assuntos
Aterosclerose , MicroRNAs , Camundongos , Animais , Músculo Liso Vascular/metabolismo , Hibridização in Situ Fluorescente , Aterosclerose/genética , Aterosclerose/metabolismo , Becaplermina/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Proliferação de Células/genética , Miócitos de Músculo Liso/metabolismo , Movimento Celular/genética , Mamíferos/metabolismo
5.
PLoS One ; 19(3): e0300370, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38536827

RESUMO

Anti-VEGF (vascular endothelial growth factor) drugs such as aflibercept (AFL) and bevacizumab (BVZ) inhibit pathological neo-angiogenesis and vascular permeability in retinal vascular diseases. As cytokines and growth factors are produced by Müller glial cells under stressful and pathological conditions, we evaluated the in vitro effect of AFL (Eylea®, 0.5 mg/mL) and BVZ (Avastin®, 0.5 mg/mL) on cell viability/metabolism, and cytokine/growth factor production by Müller cells (MIO-M1) under cobalt chloride (CoCl2)-induced hypoxia after 24h, 48h and 72h. Cell viability/metabolism were analyzed by Trypan Blue and MTT assays and cytokine/growth factors in supernatants by Luminex xMAP-based multiplex bead-based immunoassay. Cell viability increased with AFL at 48h and 72h and decreased with BVZ or hypoxia at 24h. BVZ-treated cells showed lower cell viability than AFL at all exposure times. Cell metabolism increased with AFL but decreased with BVZ (72h) and hypoxia (48h and72h). As expected, AFL and BVZ decreased VEGF levels. AFL increased PDGF-BB, IL-6 and TNF-α (24h) and BVZ increased PDGF-BB (72h). Hypoxia reduced IL-1ß, -6, -8, TNF-α and PDGF-BB at 24h, and its suppressive effect was more prominent than AFL (EGF, PDGF-BB, IL-1ß, IL-6, IL-8, and TNF-α) and BVZ (PDGF-BB and IL-6) effects. Hypoxia increased bFGF levels at 48h and 72h, even when combined with anti-VEGFs. However, the stimulatory effect of BVZ predominated over hypoxia for IL-8 and TNF-α (24h), as well as for IL-1ß (72h). Thus, AFL and BVZ exhibit distinct exposure times effects on MIO-M1 cells viability, metabolism, and cytokines/growth factors. Hypoxia and BVZ decreased MIO-M1 cell viability/metabolism, whereas AFL likely induced gliosis. Hypoxia resulted in immunosuppression, and BVZ stimulated inflammation in hypoxic MIO-M1 cells. These findings highlight the complexity of the cellular response as well as the interplay between anti-VEGF treatments and the hypoxic microenvironment.


Assuntos
Células Ependimogliais , Receptores de Fatores de Crescimento do Endotélio Vascular , Proteínas Recombinantes de Fusão , Fator A de Crescimento do Endotélio Vascular , Humanos , Bevacizumab/farmacologia , Bevacizumab/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Células Ependimogliais/metabolismo , Sobrevivência Celular , Becaplermina/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Interleucina-8/metabolismo , Interleucina-6/metabolismo , Fatores de Crescimento do Endotélio Vascular/metabolismo , Citocinas/metabolismo , Hipóxia/metabolismo , Neovascularização Patológica/patologia , Inflamação/patologia
6.
Biomolecules ; 14(3)2024 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-38540737

RESUMO

Bone morphogenetic protein (BMP) and platelet-derived growth factor (PDGF) are known to regulate/stimulate osteogenesis, playing vital roles in bone homeostasis, rendering them strong candidates for osteoporosis treatment. We evaluated the effects of recombinant human BMP-7 (rhBMP7) and PDGF-BB (rhPDGF-BB) in an oophorectomy-induced osteoporosis rat model. Forty Sprague Dawley rats underwent oophorectomy surgery; treatments commenced on the 100th day post-surgery when all animals exhibited signs of osteoporosis. These peptide growth factors were administered intraocularly (iv) once or twice a week and the animals were monitored for a total of five weeks. Two weeks after the conclusion of the treatments, the animals were euthanized and tissues were collected for assessment of alkaline phosphatase, X-ray, micro-CT, and histology. The results indicate that the most promising treatments were 20 µg/kg rhPDGF-BB + 30 µg/kg rhBMP-7 twice a week and 30 µg/kg BMP-7 twice a week, showing significant increases of 15% (p < 0.05) and 13% (p < 0.05) in bone volume fraction and 21% (p < 0.05) and 23% (p < 0.05) in trabecular number, respectively. In conclusion, rhPDGF-BB and rhBMP-7 have demonstrated the ability to increase bone volume and density in this osteoporotic animal model, establishing them as potential candidates for osteoporosis treatment.


Assuntos
Proteína Morfogenética Óssea 7 , Osteoporose , Humanos , Ratos , Animais , Becaplermina/farmacologia , Proteínas Proto-Oncogênicas c-sis/farmacologia , Proteínas Proto-Oncogênicas c-sis/uso terapêutico , Proteína Morfogenética Óssea 7/farmacologia , Proteína Morfogenética Óssea 7/uso terapêutico , Ratos Sprague-Dawley , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Proteínas Morfogenéticas Ósseas , Osteoporose/tratamento farmacológico , Proteína Morfogenética Óssea 2
7.
Biochim Biophys Acta Mol Basis Dis ; 1870(4): 167099, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38428686

RESUMO

The abnormal proliferation, migration, and inflammation of vascular smooth muscle cells (VSMCs) play crucial roles in the development of neointimal hyperplasia and restenosis. Exposure to inflammatory cytokines such as platelet-derived growth factor (PDGF)-BB and tumour necrosis factor-alpha (TNF-α) induces the transformation of contractile VSMCs into abnormal synthetic VSMCs. Isoxanthohumol (IXN) has significant anti-inflammatory, antiproliferative, and antimigratory effects. This study aimed to explore the therapeutic impact and regulatory mechanism of IXN in treating neointimal hyperplasia. The present findings indicate that IXN effectively hinders the abnormal proliferation, migration, and inflammation of VSMCs triggered by PDGF or TNF-α. This inhibition is primarily achieved through the modulation of the apelin/AKT or AKT pathway, respectively. In an in vivo model, IXN effectively reduced neointimal hyperplasia in denuded femoral arteries. These results suggest that IXN holds promise as a potential and innovative therapeutic candidate for the treatment of restenosis.


Assuntos
Proteínas Proto-Oncogênicas c-akt , Fator de Necrose Tumoral alfa , Xantonas , Humanos , Hiperplasia/tratamento farmacológico , Proliferação de Células , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Apelina , Movimento Celular , Becaplermina/farmacologia , Neointima/tratamento farmacológico , Neointima/metabolismo , Inflamação
8.
J Biochem Mol Toxicol ; 38(4): e23675, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38488158

RESUMO

Accumulating evidence shows that the abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) can significantly affect the long-term prognosis of coronary artery bypass grafting. This study aimed to explore the factors affecting the proliferation, migration, and phenotypic transformation of VSMCs. First, we stimulated VSMCs with different platelet-derived growth factor-BB (PDGF-BB) concentrations, analyzed the expression of phenotype-associated proteins by Western blotting, and examined cell proliferation by scratch wound healing and the 5-ethynyl-2-deoxyuridine (EdU) assay. VSMC proliferation was induced most by PDGF-BB treatment at 20 ng/mL. miR-200a-3p decreased significantly in A7r5 cells stimulated with PDGF-BB. The overexpression of miR-200a-3p reversed the downregulation of α-SMA (p < 0.001) and the upregulation of vimentin (p < 0.001) caused by PDGF-BB. CCK8 and EdU analyses showed that miR-200a-3p overexpression could inhibit PDGF-BB-induced cell proliferation (p < 0.001). However, flow cytometric analysis showed that it did not significantly increase cell apoptosis. Collectively, the overexpression of miR-200a-3p inhibited the proliferation and migration of VSMCs induced by PDGF-BB, partly by affecting phenotypic transformation-related proteins, providing a new strategy for relieving the restenosis of vein grafts.


Assuntos
MicroRNAs , Músculo Liso Vascular , Becaplermina/farmacologia , Proliferação de Células , Miócitos de Músculo Liso , Fenótipo , MicroRNAs/genética , Movimento Celular , Células Cultivadas
9.
Stem Cell Res Ther ; 15(1): 59, 2024 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-38433209

RESUMO

BACKGROUND: Pericytes are multifunctional contractile cells that reside on capillaries. Pericytes are critical regulators of cerebral blood flow and blood-brain barrier function, and pericyte dysfunction may contribute to the pathophysiology of human neurological diseases including Alzheimers disease, multiple sclerosis, and stroke. Induced pluripotent stem cell (iPSC)-derived pericytes (iPericytes) are a promising tool for vascular research. However, it is unclear how iPericytes functionally compare to primary human brain vascular pericytes (HBVPs). METHODS: We differentiated iPSCs into iPericytes of either the mesoderm or neural crest lineage using established protocols. We compared iPericyte and HBVP morphologies, quantified gene expression by qPCR and bulk RNA sequencing, and visualised pericyte protein markers by immunocytochemistry. To determine whether the gene expression of neural crest iPericytes, mesoderm iPericytes or HBVPs correlated with their functional characteristics in vitro, we quantified EdU incorporation following exposure to the key pericyte mitogen, platelet derived growth factor (PDGF)-BB and, contraction and relaxation in response to the vasoconstrictor endothelin-1 or vasodilator adenosine, respectively. RESULTS: iPericytes were morphologically similar to HBVPs and expressed canonical pericyte markers. However, iPericytes had 1864 differentially expressed genes compared to HBVPs, while there were 797 genes differentially expressed between neural crest and mesoderm iPericytes. Consistent with the ability of HBVPs to respond to PDGF-BB signalling, PDGF-BB enhanced and a PDGF receptor-beta inhibitor impaired iPericyte proliferation. Administration of endothelin-1 led to iPericyte contraction and adenosine led to iPericyte relaxation, of a magnitude similar to the response evoked in HBVPs. We determined that neural crest iPericytes were less susceptible to PDGFR beta inhibition, but responded most robustly to vasoconstrictive mediators. CONCLUSIONS: iPericytes express pericyte-associated genes and proteins and, exhibit an appropriate physiological response upon exposure to a key endogenous mitogen or vasoactive mediators. Therefore, the generation of functional iPericytes would be suitable for use in future investigations exploring pericyte function or dysfunction in neurological diseases.


Assuntos
Células-Tronco Pluripotentes Induzidas , Pericitos , Humanos , Becaplermina/farmacologia , Endotelina-1/farmacologia , Adenosina , Proliferação de Células
10.
Mol Immunol ; 168: 38-46, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38422885

RESUMO

Asthma, a common pediatric pulmonary disease, significantly affects children's healthy development. This study aimed to investigate the functions of human ß defensin-3 (HBD-3) in asthma progression. For this purpose, blood samples from asthmatic and healthy children were collected. Moreover, the airway smooth muscle cells (ASMCs) were treated with platelet-derived growth factor BB (PDGF-BB) to develop an in vitro asthma model, then evaluated cell viability and migration via CCK-8 and transwell assays. The mRNA levels of interferon γ (INF-γ), interleukin 4 (IL-4), interleukin 10 (IL-10), alpha-smooth muscle actin (α-SMA), HBD-3, and the protein levels of phosphatidylinositol 3-kinase (PI3K) along with protein kinase B (AKT) were detected. Similarly, the N6-methyladenosine (m6A) content in the ASMCs and m6A levels of HBD-3 were also measured. Results indicated an upregulated HBD-3 in the asthmatic children. The ASMCs were found to be stimulated by PDGF-BB, in addition to the promotion of cell viability and migration. The INF-γ, IL-4, and α-SMA levels were reduced, while IL-10 was elevated in PDGF-BB-stimulated ASMCs. Silencing HBD-3 in PDGF-BB stimulated ASMCs was found to exert the opposite effect by inhibiting cell viability and migration, enhancing the levels of INF-γ, IL-4, and α-SMA, while the IL-10 levels were found to decline. PDGF-BB stimulation of ASMCs resulted in activation of the PI3K/AKT signaling pathway, which was blocked post HBD-3 silencing, while the role of si-hBD in PDGF-BB stimulated ASMCs was neutralized post-treatment with IGF-1. Finally, it was found that METTL3 overexpression prominently upregulated the m6A levels of HBD-3 and decreased the mRNA expression and stability of HBD-3 in the PDGF-BB-stimulated ASMCs. The study concluded that METTL3-mediated HBD-3 participates in the progression of asthma through the PI3K/AKT signaling pathway.


Assuntos
Asma , Metiltransferases , Miócitos de Músculo Liso , beta-Defensinas , Criança , Humanos , Asma/metabolismo , Becaplermina/farmacologia , Becaplermina/metabolismo , beta-Defensinas/genética , beta-Defensinas/metabolismo , Movimento Celular/genética , Proliferação de Células/genética , Células Cultivadas , Interleucina-10/metabolismo , Interleucina-4/metabolismo , Pulmão/metabolismo , Miócitos de Músculo Liso/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/metabolismo , Transdução de Sinais
11.
Cardiovasc Toxicol ; 24(2): 111-121, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38378972

RESUMO

Circular RNA (circRNAs) has been confirmed to participate in atherosclerosis (AS) progression. However, the role and mechanism of hsa_circ_0032389 in AS process still need to be further revealed. This study evaluates the role and mechanism of hsa_circ_0032389 in AS process. Platelet-derived growth factor-BB (PDGF-BB) was used to induce human aortic vascular smooth muscle cells (HA-VSMCs). The expression levels of hsa_circ_0032389, microRNA (miR)-513a-5p, and fibroblast growth factor receptor substrate 2 (FRS2) were examined by quantitative real-time PCR. Cell proliferation and migration were analyzed using cell counting kit 8 assay, flow cytometry, EdU assay, transwell assay, and wound healing assay. Protein expression was assessed using western blot analysis. Dual-luciferase reporter and RIP assays were used to confirm RNA interaction. Hsa_circ_0032389 was overexpressed in PDGF-BB-induced HA-VSMCs, and its downregulation inhibited HA-VSMC viability, cell cycle, EdU positive cell rate, migratory cell number, and wound closure rate under PDGF-BB treatment. The luciferase activity of hsa_circ_0032389wt could be reduced by miR-513a-5p mimic, and both hsa_circ_0032389 and miR-513a-5p were enriched in anti-Ago2, confirming that miR-513a-5p could be sponged by hsa_circ_0032389. MiR-513a-5p inhibitor reversed the effect of hsa_circ_0032389 knockdown on PDGF-BB-induced HA-VSMC viability, cell cycle, EdU positive cell rate, migratory cell number, and wound closure rate. Moreover, the luciferase activity of FRS2wt was reduced by miR-513a-5p mimic, and both FRS2 and miR-513a-5p were enriched in anti-Ago2, verifying that FRS2 was targeted by miR-513a-5p. MiR-513a-5p suppressed PDGF-BB-induced HA-VSMC viability, cell cycle, EdU positive cell rate, migratory cell number, and wound closure rate by targeting FRS2. Our results indicated that hsa_circ_0032389 enhanced PDGF-BB-induced HA-VSMC proliferation and migration via regulating miR-513a-5p/FRS2 axis.


Assuntos
Aterosclerose , MicroRNAs , Humanos , Becaplermina/farmacologia , Músculo Liso Vascular , MicroRNAs/genética , Proliferação de Células , Luciferases , Movimento Celular
12.
J Agric Food Chem ; 72(8): 4008-4022, 2024 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-38373191

RESUMO

The vital pathological processes in intimal hyperplasia include aberrant vascular smooth muscle cells (VSMCs) proliferation, migration, and phenotypic switching. Rosmarinic acid (RA) is a natural phenolic acid compound. Nevertheless, the underlying mechanism of RA in neointimal hyperplasia is still unclear. Our analysis illustrated that miR-25-3p mimics significantly enhanced PDGF-BB-mediated VSMCs proliferation, migration, and phenotypic switching while RA partially weakened the effect of miR-25-3p. Mechanistically, we found that miR-25-3p directly targets sirtuin (SIRT6). The suppressive effect of the miR-25-3p inhibitor on PDGF-BB-induced VSMCs proliferation, migration, and phenotypic switch was partially eliminated by SIRT6 knockdown. The suppression of the PDGF-BB-stimulated Nrf2/ARE signaling pathway that was activated by the miR-25-3p inhibitor was exacerbated by the SIRT6 knockdown. In in vivo experiments, RA reduced the degree of intimal hyperplasia while miR-25-3p agomir partially reversed the suppressive effect of RA in vascular remodeling. Our results indicate that RA activates the Nrf2/ARE signaling pathway via the miR-25-3p/SIRT6 axis to inhibit vascular remodeling.


Assuntos
MicroRNAs , Sirtuínas , Humanos , Becaplermina/farmacologia , Proliferação de Células , Hiperplasia/metabolismo , Hiperplasia/patologia , 60556 , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Remodelação Vascular , Músculo Liso Vascular , Movimento Celular , Transdução de Sinais , MicroRNAs/genética , MicroRNAs/metabolismo , Miócitos de Músculo Liso , Células Cultivadas , Sirtuínas/metabolismo , Sirtuínas/farmacologia
13.
Discov Med ; 36(181): 323-331, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38409837

RESUMO

BACKGROUND: Childhood asthma is a chronic inflammatory disease of the respiratory tract characterized by bronchial inflammation, airway hyperresponsiveness, airflow disorder, and obstruction. Secreted frizzled-related protein 5 (SFRP5) may be associated with respiratory inflammatory diseases. This study investigated the effect of SFRP5 on human airway smooth muscle cells (HASMCs) to provide new ideas for treating asthma. METHODS: A total of 30 children with asthma and 30 children who had a physical examination at the same time were selected and divided into asthma and healthy groups. Serum SFRP5 levels were determined by enzyme-linked immunosorbent assay (ELISA) and real-time quantitative polymerase chain reaction (RT-qPCR). Lipofectamine 2000™ regent was used to transfect the SFRP5 overexpression plasmid (pc-SFRP5) or corresponding negative control (pc-NC) into HASMCs. HASMCs were treated with 10 µg/L platelet-derived growth factor-BB (PDGF-BB), which is an inducer to mimic the asthma-like condition at the cellular level of childhood asthma. HASMCs were divided into control, PDGF-BB (PDGF-BB treatment), PDGF-BB+pc-NC (pc-NC transfection and PDGF-BB treatment), and PDGF-BB+pc-SFRP5 (pc-SFRP5 transfection and PDGF-BB treatment) groups. Cell proliferation was measured by 5-ethynyl-2'-deoxyuridine (EdU) and cell counting kit-8 (CCK-8) assay. Cell migration was detected by Transwell assay. The protein expression was detected by western blot. RESULTS: Serum SFRP5 expression in the asthmatic group was decreased versus the healthy group (p < 0.0001). Induction of PDGF-BB decreased SFRP5 expression in HASMCs (p < 0.01). SFRP5 expression in the pc-SFRP5 group was increased (p < 0.01). The proliferation and migration of HASMCs increased after PDGF-BB treatment (p < 0.001, p < 0.0001), indicating that the asthma model was successfully inducted in vitro. Moreover, the expression of ß-catenin, cellular-myelocytomatosis viral oncogene (c-Myc), and cyclinD1 proteins in HASMCs increased after PDGF-BB treatment (p < 0.0001). SFRP5 overexpression partly inhibited PDGF-BB-induced proliferation, migration, and expressions of ß-catenin, c-Myc, and cyclinD proteins in HASMCs (p < 0.01, p < 0.001, p < 0.0001). CONCLUSIONS: Serum SFRP5 expression decreases in children with asthma. SFRP5 overexpression partially inhibits PDGF-BB-induced HASMC proliferation and migration by regulating the wingless-type mouse mammary tumor virus (MMTV) integration site family (Wnt)/ß-catenin pathway.


Assuntos
Asma , beta Catenina , Animais , Criança , Camundongos , Humanos , Becaplermina/metabolismo , Becaplermina/farmacologia , beta Catenina/metabolismo , beta Catenina/farmacologia , Via de Sinalização Wnt/genética , Asma/genética , Asma/metabolismo , Asma/patologia , Proliferação de Células/genética , Pulmão/metabolismo , Movimento Celular , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Células Cultivadas , Proteínas Adaptadoras de Transdução de Sinal/metabolismo
14.
Atherosclerosis ; 390: 117470, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38342025

RESUMO

BACKGROUND AND AIMS: Myotubularin-related protein 7 (MTMR7) suppresses proliferation in various cell types and is associated with cardiovascular and cerebrovascular diseases. However, whether MTMR7 regulates vascular smooth muscle cell (VSMC) and vascular intimal hyperplasia remains unclear. We explored the role of MTMR7 in phenotypic switching of VSMC and vascular intimal hyperplasia after injury. METHODS AND RESULTS: MTMR7 expression was significantly downregulated in injured arteries. Compared to wild type (WT) mice, Mtmr7-transgenic (Mtmr7-Tg) mice showed reduced intima/media ratio, decreased percentage of Ki-67-positive cells within neointima, and increased Calponin expression in injured artery. In vitro, upregulating MTMR7 by Len-Mtmr7 transfection inhibited platelet derived growth factor (PDGF)-BB-induced proliferation, migration of VSMC and reversed PDGF-BB-induced decrease in expression of Calponin and SM-MHC. Microarray, single cell sequence, and other bioinformatics analysis revealed that MTMR7 is highly related to glucose metabolism and mammalian target of rapamycin complex 1 (mTORC1). Further experiments confirmed that MTMR7 markedly repressed glycolysis and mTORC1 activity in PDGF-BB-challenged VSMC in vitro. Restoring mTORC1 activity abolished MTMR7-mediated suppression of glycolysis, phenotypic shift in VSMC in vitro and protection against vascular intimal hyperplasia in vivo. Furthermore, upregulating MTMR7 in vitro led to dephosphorylation and dissociation of p62 from mTORC1 in VSMC. External expression of p62 in vitro also abrogated the inhibitory effects of MTMR7 on glycolysis and phenotypic switching in PDGF-BB-stimulated VSMC. CONCLUSIONS: Our study demonstrates that MTMR7 inhibits injury-induced vascular intimal hyperplasia and phenotypic switching of VSMC. Mechanistically, the beneficial effects of MTMR7 are conducted via suppressing p62/mTORC1-mediated glycolysis.


Assuntos
Músculo Liso Vascular , Neointima , Camundongos , Animais , Becaplermina/farmacologia , Becaplermina/metabolismo , Proliferação de Células , Músculo Liso Vascular/patologia , Hiperplasia/patologia , Neointima/metabolismo , Camundongos Transgênicos , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/farmacologia , Glucose/metabolismo , Miócitos de Músculo Liso/patologia , Movimento Celular , Células Cultivadas , Mamíferos
15.
Mol Med ; 30(1): 21, 2024 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-38317079

RESUMO

BACKGROUND: Pericytes are a vital component of the blood-brain barrier, and their involvement in acute inflammation was recently suggested. However, it remains unclear whether pericytes contribute to hypothalamic chronic inflammation and energy metabolism in obesity. The present study investigated the impact of pericytes on the pathophysiology of obesity by focusing on platelet-derived growth factor (PDGF) signaling, which regulates pericyte functions. METHODS: Tamoxifen-inducible systemic conditional PDGF receptor ß knockout mice (Pdgfrb∆SYS-KO) and Calcium/calmodulin-dependent protein kinase type IIa (CaMKIIa)-positive neuron-specific PDGF receptor ß knockout mice (Pdgfrb∆CaMKII-KO) were fed a high-fat diet, and metabolic phenotypes before and 3 to 4 weeks after dietary loading were examined. Intracellular energy metabolism and relevant signal transduction in lipopolysaccharide- and/or platelet-derived growth factor-BB (PDGF-BB)-stimulated human brain pericytes (HBPCs) were assessed by the Seahorse XFe24 Analyzer and Western blotting. The pericyte secretome in conditioned medium from HBPCs was studied using cytokine array kit, and its impact on polarization was examined in bone marrow-derived macrophages (BMDMs), which are microglia-like cells. RESULTS: Energy consumption increased and body weight gain decreased after high-fat diet loading in Pdgfrb∆SYS-KO mice. Cellular oncogene fos (cFos) expression increased in proopiomelanocortin (POMC) neurons, whereas microglial numbers and inflammatory gene expression decreased in the hypothalamus of Pdgfrb∆SYS-KO mice. No significant changes were observed in Pdgfrb∆CaMKII-KO mice. In HBPCs, a co-stimulation with lipopolysaccharide and PDGF-BB shifted intracellular metabolism towards glycolysis, activated mitogen-activated protein kinase (MAPK), and modulated the secretome to the inflammatory phenotype. Consequently, the secretome showed an increase in various proinflammatory chemokines and growth factors including Epithelial-derived neutrophil-activating peptide 78 (C-X-C motif chemokine ligand (CXCL)5), Thymus and activation-regulated chemokine (C-C motif chemokine (CCL)17), Monocyte chemoattractant protein 1 (CCL2), and Growth-regulated oncogene α (CXCL1). Furthermore, conditioned medium from HBPCs stimulated the inflammatory priming of BMDMs, and this change was abolished by the C-X-C motif chemokine receptor (CXCR) inhibitor. Consistently, mRNA expression of CXCL5 was elevated by lipopolysaccharide and PDGF-BB treatment in HBPCs, and the expression was significantly lower in the hypothalamus of Pdgfrb∆SYS-KO mice than in control Pdgfrbflox/flox mice (FL) following 4 weeks of HFD feeding. CONCLUSIONS: PDGF receptor ß signaling in hypothalamic pericytes promotes polarization of macrophages by changing their secretome and contributes to the progression of obesity.


Assuntos
Pericitos , Fator de Crescimento Derivado de Plaquetas , Camundongos , Humanos , Animais , Fator de Crescimento Derivado de Plaquetas/metabolismo , Fator de Crescimento Derivado de Plaquetas/farmacologia , Pericitos/metabolismo , Becaplermina/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Meios de Cultivo Condicionados/metabolismo , Lipopolissacarídeos , Transdução de Sinais , Inflamação/metabolismo , Camundongos Knockout , Obesidade/metabolismo , Hipotálamo , Proteínas Proto-Oncogênicas c-sis/genética , Proteínas Proto-Oncogênicas c-sis/metabolismo
16.
Eur J Pharmacol ; 968: 176422, 2024 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-38365108

RESUMO

Vascular smooth muscle cells (VSMCs) contribute to neointimal hyperplasia (NIH) after vascular injury, a common feature of vascular remodelling disorders. Suramin is known to exert antitumour effects by inhibiting the proliferation of various tumour cells; however, its effects and mechanism on VSMCs remain unclear. This study investigated the effects of suramin on human aortic smooth muscle cells (HASMCs), rat aortic smooth muscle cells (RASMCs) and NIH to examine its suitability for the prevention of vascular remodelling disorders. In vitro, suramin administration reduced platelet-derived growth factor type BB (PDGF-BB)-stimulated proliferation, migration, and dedifferentiation of VSMCs through a transforming growth factor beta receptor 1 (TGFBR1)/Smad2/3-dependent pathway. Suramin dramatically inhibited NIH ligation in the left common carotid artery (LCCA) vivo. Therefore, our results indicate that suramin protects against the development of pathological vascular remodelling by attenuating VSMCs proliferation, migration, and phenotypic transformation and may be used as a potential medicine for the treatment of NIH.


Assuntos
Neointima , Suramina , Ratos , Humanos , Animais , Hiperplasia/patologia , Proliferação de Células , Suramina/farmacologia , Suramina/metabolismo , Neointima/patologia , Músculo Liso Vascular , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Remodelação Vascular , Becaplermina/farmacologia , Miócitos de Músculo Liso , Movimento Celular , Células Cultivadas
17.
Sci Rep ; 14(1): 4191, 2024 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-38378786

RESUMO

Lung adenocarcinoma (LUAD) remains one of the most aggressive tumors and the efficacy of conventional treatment has been bleak. Nowadays, gene-targeted therapy has become a new favorite in tumor therapy. Herein, we investigated the effect of platelet derived growth factor BB (PDGFBB) on LUAD. Firstly, PDGFBB was upregulated in LUAD patients and closely linked with poor survival. Furthermore, the expression of PDGFBB and PDGFRα/ß in LUAD cells was higher than that in normal lung cells. By loss-of-function with herpes simplex virus (HSV)-PDGFi-shRNA, we found that PDGFBB knockdown caused a significant decrease in proliferation and migration, but evoked apoptosis of LUAD cells in vitro. Conversely, exogenous PDGFBB held adverse effect. Additionally, A549 cells with PDGFBB knockdown had a low probability of tumorigenesis in vivo. Moreover, PDGFBB knockdown restrained the growth of xenografts derived from normal A549 cells. Mechanistically, PDGFBB knockdown suppressed PI3K/AKT and Ras/MAPK signaling, while PDGFBB was the opposite. Therefore, we concluded that PDGFBB might facilitate the tumorigenesis and malignancy of LUAD through its functional downstream nodes-PI3K/AKT and Ras/MAPK signaling, which supported that PDGFBB could serve as a rational therapeutic target for LUAD.


Assuntos
Adenocarcinoma de Pulmão , Neoplasias Pulmonares , Humanos , Becaplermina/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Neoplasias Pulmonares/patologia , Adenocarcinoma de Pulmão/patologia , Transformação Celular Neoplásica/genética , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Movimento Celular/genética , Linhagem Celular Tumoral
18.
Cardiovasc Toxicol ; 24(3): 225-239, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38324114

RESUMO

Aortic dissection (AD) is a severe vascular disease with high rates of mortality and morbidity. However, the underlying molecular mechanisms of AD remain unclear. Differentially expressed genes (DEGs) were screened by bioinformatics methods. Alterations of histopathology and inflammatory factor levels in ß-aminopropionitrile (BAPN)-induced AD mouse model were evaluated through Hematoxylin-Eosin (HE) staining and Enzyme-linked immunosorbent assay (ELISA), respectively. Reverse transcription quantitative real-time polymerase chain reaction was performed to detect DEGs expression. Furthermore, the role of LILRB4 in AD was investigated through Cell Counting Kit-8 (CCK-8), wound healing, and flow cytometry. Western blotting was employed to assess the phenotypic switch and extracellular matrix (ECM)-associated protein expressions in platelet-derived growth factor-BB (PDGF-BB)-stimulated in vitro model of AD. In the AD mouse model, distinct dissection formation was observed. TNF-α, IL-1ß, IL-8, and IL-6 levels were higher in the AD mouse model than in the controls. Six hub genes were identified, including LILRB4, TIMP1, CCR5, CCL7, MSR1, and CLEC4D, all of which were highly expressed. Further exploration revealed that LILRB4 knockdown inhibited the cell vitality and migration of PDGF-BB-induced HASMCs while promoting apoptosis and G0/G1 phase ratio. More importantly, LILRB4 knockdown promoted the protein expression of α-SMA and SM22α, while decreasing the expression of Co1, MMP2, and CTGF, which suggested that LILRB4 silencing promoted contractile phenotypic transition and ECM stability. LILRB4 knockdown inhibits the progression of AD. Our study provides a new potential target for the clinical treatment of AD.


Assuntos
Dissecção Aórtica , Camundongos , Animais , Humanos , Becaplermina/metabolismo , Becaplermina/farmacologia , Regulação para Baixo , Miócitos de Músculo Liso/metabolismo , Apoptose , Proliferação de Células , Movimento Celular , Células Cultivadas , Glicoproteínas de Membrana/metabolismo , Receptores Imunológicos/metabolismo
19.
Atherosclerosis ; 390: 117430, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38301602

RESUMO

BACKGROUND AND AIMS: Tripartite motif (TRIM65) is an important member of the TRIM protein family, which is a newly discovered E3 ligase that interacts with and ubiquitinates various substrates and is involved in diverse pathological processes. However, the function of TRIM65 in atherosclerosis remains unarticulated. In this study, we investigated the role of TRIM65 in the pathogenesis of atherosclerosis, specifically in vascular smooth muscle cells (VSMCs) phenotype transformation, which plays a crucial role in formation of atherosclerotic lesions. METHODS AND RESULTS: Both non-atherosclerotic and atherosclerotic lesions during autopsy were collected singly or pairwise from each individual (n = 16) to investigate the relationship between TRIM65 and the development of atherosclerosis. In vivo, Western diet-fed ApoE-/- mice overexpressing or lacking TRIM65 were used to assess the physiological function of TRIM65 on VSMCs phenotype, proliferation and atherosclerotic lesion formation. In vitro, VSMCs phenotypic transformation was induced by platelet-derived growth factor-BB (PDGF-BB). TRIM65-overexpressing or TRIM65-abrogated primary mouse aortic smooth muscle cells (MOASMCs) and human aortic smooth muscle cells (HASMCs) were used to investigate the mechanisms underlying the progression of VSMCs phenotypic transformation, proliferation and migration. Increased TRIM65 expression was detected in α-SMA-positive cells in the medial and atherosclerotic lesions of autopsy specimens. TRIM65 overexpression increased, whereas genetic knockdown of TRIM65 remarkably inhibited, atherosclerotic plaque development. Mechanistically, TRIM65 overexpression activated PI3K/Akt/mTOR signaling, resulting in the loss of the VSMCs contractile phenotype, including calponin, α-SMA, and SM22α, as well as cell proliferation and migration. However, opposite phenomena were observed when TRIM65 was deficient in vivo or in vitro. Moreover, in cultured PDGF-BB-induced TRIM65-overexpressing VSMCs, inhibition of PI3K by treatment with the inhibitor LY-294002 for 24 h markedly attenuated PI3K/Akt/mTOR activation, regained the VSMCs contractile phenotype, and blocked the progression of cell proliferation and migration. CONCLUSIONS: TRIM65 overexpression enhances atherosclerosis development by promoting phenotypic transformation of VSMCs from contractile to synthetic state through activation of the PI3K/Akt/mTOR signal pathway.


Assuntos
Aterosclerose , Proteínas Proto-Oncogênicas c-akt , Humanos , Camundongos , Animais , Becaplermina/genética , Becaplermina/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Músculo Liso Vascular/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Movimento Celular , Transdução de Sinais , Proliferação de Células , Serina-Treonina Quinases TOR/metabolismo , Aterosclerose/patologia , Miócitos de Músculo Liso/patologia , Fenótipo , Células Cultivadas , Proteínas com Motivo Tripartido/genética , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/genética
20.
Res Vet Sci ; 168: 105138, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38218061

RESUMO

Autologous platelet-rich plasma (PRP) contains growth factors (GFs) that modulate the expression of inflammatory cells; thus, these products could be considered a good strategy to favor tissue regeneration in feline immunodeficiency (FIV) positive cats. However, there is no scientific documentation on obtaining PRP in FIV-positive cats. Authors hypothesized that PRP can be obtained in FIV cats following the PRGF®-Endoret® methodology. The objectives of this study were to compare the platelet, erythrocyte, and leukocyte concentration between whole blood (WB) and the PRP; and determine the concentration of platelet-derived growth factor BB (PDGF-BB) and transforming growth factor ß1 (TGF-ß1) in FIV-positive cats. Sixteen adults FIV-positive asymptomatic cats were included in the study. WB samples were drawn and the PRP was obtained by centrifugation at 265g for 10 min. Erythrocyte and leukocyte, platelets, and mean platelet volume (MPV) were determined both in WB and in PRP. PDGF-BB and TGF-ß1 concentrations were additionally determined in PRP. Platelet concentration increased 1.1 times in PRP fraction compared to WB, but no significant differences were reported. MPV was statistically higher in WB than in PRP (p = 0.001). Erythrocytes and leukocytes counts were decreased by 99% and 92%, respectively in the PRP fraction (p < 0.001). Regarding TGF-ß1, a higher concentration was shown in the PRP (p < 0.02). Although the product obtained could not be classified as PRP according to the PRGF®-Endoret® methodology, based on the drastic reduction of RBC and WBC, the PLT concentrate is of high purity.


Assuntos
Vírus da Imunodeficiência Felina , Plasma Rico em Plaquetas , Gatos , Animais , Becaplermina/metabolismo , Fator de Crescimento Transformador beta1/análise , Fator de Crescimento Transformador beta1/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Plaquetas , Plasma Rico em Plaquetas/química , Plasma Rico em Plaquetas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...