Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
Cell Mol Biol Lett ; 29(1): 47, 2024 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-38589823

RESUMEN

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.


Asunto(s)
Glucosafosfato Deshidrogenasa , Músculo Liso Vascular , Canal Aniónico 1 Dependiente del Voltaje , Humanos , Hiperplasia/metabolismo , Hiperplasia/patología , Becaplermina/genética , Becaplermina/metabolismo , Proliferación Celular , Proteína X Asociada a bcl-2/metabolismo , Glucosafosfato Deshidrogenasa/metabolismo , Músculo Liso Vascular/metabolismo , Cromatografía Liquida , Espectrometría de Masas en Tándem , Neointima/genética , Neointima/metabolismo , Neointima/patología , Apoptosis , Miocitos del Músculo Liso/metabolismo , Movimiento Celular/genética , Células Cultivadas , Fenotipo
2.
Atherosclerosis ; 390: 117430, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38301602

RESUMEN

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.


Asunto(s)
Aterosclerosis , Proteínas Proto-Oncogénicas c-akt , Humanos , Ratones , Animales , Becaplermina/genética , Becaplermina/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Músculo Liso Vascular/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Movimiento Celular , Transducción de Señal , Proliferación Celular , Serina-Treonina Quinasas TOR/metabolismo , Aterosclerosis/patología , Miocitos del Músculo Liso/patología , Fenotipo , Células Cultivadas , Proteínas de Motivos Tripartitos/genética , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/genética
3.
Cell Biochem Funct ; 42(1): e3896, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38081793

RESUMEN

Cysteine and glycine-rich protein 2 (Csrp2) has emerged as a key factor in controlling the phenotypic modulation of smooth muscle cells. The phenotypic transition of airway smooth muscle cells (ASMCs) is a pivotal step in developing airway remodeling during the onset of asthma. However, whether Csrp2 mediates the phenotypic transition of ASMCs in airway remodeling during asthma onset is undetermined. This work aimed to address the link between Csrp2 and the phenotypic transition of ASMCs evoked by platelet-derived growth factor (PDGF)-BB in vitro. The overexpression or silencing of Csrp2 in ASMCs was achieved through adenovirus-mediated gene transfer. The expression of mRNA was measured by quantitative real-time-PCR. Protein levels were determined through Western blot analysis. Cell proliferation was detected by EdU assay and Calcein AM assays. Cell cycle distribution was assessed via fluorescence-activated cell sorting assay. Cell migration was evaluated using the scratch-wound assay. The transcriptional activity of Yes-associated protein (YAP)/transcriptional coactivator with PDZ-binding motif (TAZ) was measured using the luciferase reporter assay. A decline in Csrp2 level occurred in PDGF-BB-stimulated ASMCs. Increasing Csrp2 expression repressed the PDGF-BB-evoked proliferation and migration of ASMCs. Moreover, increasing Csrp2 expression impeded the phenotypic change of PDGF-BB-stimulated ASMCs from a contractile phenotype into a synthetic/proliferative phenotype. On the contrary, the opposite effects were observed in Csrp2-silenced ASMCs. The activity of YAP/TAZ was elevated in PDGF-BB-stimulated ASMCs, which was weakened by Csrp2 overexpression or enhanced by Csrp2 silencing. The YAP/TAZ activator could reverse Csrp2-overexpression-mediated suppression of the PDGF-BB-evoked phenotypic switching of ASMCs, while the YAP/TAZ suppressor could dimmish Csrp2-silencing-mediated enhancement on PDGF-BB-evoked phenotypic switching of ASMCs. In summary, Csrp2 serves as a determinant for the phenotypic switching of ASMCs. Increasing Csrp2 is able to impede PDGF-BB-evoked phenotypic change of ASMCs from a synthetic phenotype into a synthetic/proliferative phenotype through the effects on YAP/TAZ. This work implies that Csrp2 may be a key player in airway remodeling during the onset of asthma.


Asunto(s)
Asma , Cisteína , Humanos , Becaplermina/genética , Becaplermina/metabolismo , Cisteína/genética , Cisteína/metabolismo , Remodelación de las Vías Aéreas (Respiratorias) , Células Cultivadas , Miocitos del Músculo Liso/metabolismo , Proliferación Celular , Asma/metabolismo , Fenotipo , Movimiento Celular
4.
J Clin Invest ; 133(23)2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-37815871

RESUMEN

Brain vascular calcification is a prevalent age-related condition often accompanying neurodegenerative and neuroinflammatory diseases. The pathogenesis of large-vessel calcifications in peripheral tissue is well studied, but microvascular calcification in the brain remains poorly understood. Here, we report that elevated platelet-derived growth factor BB (PDGF-BB) from bone preosteoclasts contributed to cerebrovascular calcification in male mice. Aged male mice had higher serum PDGF-BB levels and a higher incidence of brain calcification compared with young mice, mainly in the thalamus. Transgenic mice with preosteoclast-specific Pdgfb overexpression exhibited elevated serum PDGF-BB levels and recapitulated age-associated thalamic calcification. Conversely, mice with preosteoclast-specific Pdgfb deletion displayed diminished age-associated thalamic calcification. In an ex vivo cerebral microvascular culture system, PDGF-BB dose-dependently promoted vascular calcification. Analysis of osteogenic gene array and single-cell RNA-Seq (scRNA-Seq) revealed that PDGF-BB upregulated multiple osteogenic differentiation genes and the phosphate transporter Slc20a1 in cerebral microvessels. Mechanistically, PDGF-BB stimulated the phosphorylation of its receptor PDGFRß (p-PDGFRß) and ERK (p-ERK), leading to the activation of RUNX2. This activation, in turn, induced the transcription of osteoblast differentiation genes in PCs and upregulated Slc20a1 in astrocytes. Thus, bone-derived PDGF-BB induced brain vascular calcification by activating the p-PDGFRß/p-ERK/RUNX2 signaling cascade in cerebrovascular cells.


Asunto(s)
Subunidad alfa 1 del Factor de Unión al Sitio Principal , Calcificación Vascular , Masculino , Ratones , Animales , Becaplermina/genética , Becaplermina/metabolismo , Proteínas Proto-Oncogénicas c-sis/genética , Proteínas Proto-Oncogénicas c-sis/metabolismo , Proteínas Proto-Oncogénicas c-sis/farmacología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Osteogénesis , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Encéfalo/metabolismo , Calcificación Vascular/genética
5.
Epigenetics ; 18(1): 2242225, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37537976

RESUMEN

Idiopathic pulmonary arterial hypertension (IPAH) is a serious and fatal disease. Recently, m6A has been reported to play an important role in the lungs of IPAH patients and experimental pulmonary hypertension models. However, the meaning of m6A mRNAs in the peripheral blood of IPAH patients remains largely unexplored. We aimed to construct a transcriptome-wide map of m6A mRNAs in the peripheral blood of IPAH patients. M6A RNA Methylation Quantification Kit was utilized to measure the total m6A levels in the peripheral blood of IPAH patients. A combination of MeRIP-seq, RNA-seq and bioinformatics analysis was utilized to select m6A-modified hub genes of IPAH. MeRIP-qPCR and RT-qPCR were used to measure the m6A levels and mRNA levels of TP53, RPS27A, SMAD3 and FoxO3 in IPAH patients. Western blot was performed to assess the protein levels of m6A related regulators and m6A related genes in experimental PH animal models, hypoxia-treated and PDGF-BB induced PASMCs. We found that the total m6A levels were increased in peripheral blood of IPAH patients and verified that m6A levels of RPS27A and SMAD3 were significantly elevated and m6A levels of TP53 and FoxO3 were significantly reduced. The mRNA or protein levels of RPS27A, SMAD3, TP53 and FoxO3 were changed in human blood samples, experimental PH animal models and PDGF-BB induced PASMCs. Moreover, METTL3 and YTHDF1 were increased in the hypoxia induced pulmonary hypertension rat model, hypoxia-treated and PDGF-BB induced PASMCs. These finding suggested that m6A may play an important role in IPAH.


Asunto(s)
Hipertensión Pulmonar , Humanos , Ratas , Animales , Hipertensión Pulmonar Primaria Familiar/genética , Hipertensión Pulmonar Primaria Familiar/metabolismo , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Becaplermina/genética , Becaplermina/metabolismo , Arteria Pulmonar/metabolismo , Epigenoma , Proliferación Celular , Miocitos del Músculo Liso/metabolismo , Metilación de ADN , ARN Mensajero/metabolismo , Metiltransferasas/genética , Metiltransferasas/metabolismo
6.
Sci Rep ; 13(1): 10422, 2023 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-37369741

RESUMEN

Soft tissue sarcomas (STS) are a heterogenous group of mesenchymal tumors representing over 50 distinct types with overlapping histological features and non-specific anatomical locations. Currently, localized sarcomas are treated with surgery + / - radiation in both humans and dogs with few molecularly targeted therapeutic options. However, to improve precision-based cancer therapy through trials in pet dogs with naturally occurring STS tumors, knowledge of genomic profiling and molecular drivers in both species is essential. To this purpose, we sought to characterize the transcriptomic and genomic mutation profiles of canine STS subtypes (fibrosarcoma, undifferentiated pleomorphic sarcoma, and peripheral nerve sheath tumors), by leveraging RNAseq, whole exome sequencing, immunohistochemistry, and drug assays. The most common driver mutations were in cell cycle/DNA repair (31%, TP53-21%) and chromatin organization/binding (41%, KMT2D-21%) genes. Similar to a subset of human sarcomas, we identified fusion transcripts of platelet derived growth factor B and collagen genes that predict sensitivity to PDGFR inhibitors. Transcriptomic profiling grouped these canine STS tumors into 4 clusters, one PNST group (H1), and 3 FSA groups selectively enriched for extracellular matrix interactions and PDFGB fusions (H2), homeobox transcription factors (H3), and elevated T-cell infiltration (H4). This multi-omics approach provides insights into canine STS sub-types at a molecular level for comparison to their human counterparts, to improve diagnosis, and may provide additional targets for chemo- and immuno-therapy.


Asunto(s)
Histiocitoma Fibroso Maligno , Sarcoma , Neoplasias de los Tejidos Blandos , Animales , Perros , Becaplermina/genética , Mutación , Proteínas Proto-Oncogénicas c-sis/genética , Sarcoma/genética , Sarcoma/veterinaria , Sarcoma/patología , Neoplasias de los Tejidos Blandos/patología , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/genética
7.
Stem Cell Rev Rep ; 19(5): 1554-1575, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37060532

RESUMEN

Mesenchymal stem cells (MSCs) is promising in promoting wound healing mainly due to their paracrine function. Nonetheless, the transplanted MSCs presented poor survival with cell dysfunction and paracrine problem in diabetic environment, thus limiting their therapeutic efficacy and clinical application. JAM-A, an adhesion molecule, has been reported to play multi-functional roles in diverse cells. We therefore investigated the potential effect of JAM-A on MSCs under diabetic environment and explored the underlying mechanism. Indeed, high-glucose condition inhibited MSCs viability and JAM-A expression. However, JAM-A abnormality was rescued by lentivirus transfection and JAM-A overexpression promoted MSCs proliferation, migration and adhesion under hyperglycemia. Moreover, JAM-A overexpression attenuated high-glucose-induced ROS production and MSCs apoptosis. The bio-effects of JAM-A on MSCs under hyperglycemia were confirmed by RNA-seq with enrichment analyses. Moreover, Luminex chip results showed JAM-A overexpression dramatically upregulated PDGF-BB and VEGF in the supernatant of MSCs, which was verified by RT-qPCR and western blotting. The supernatant was further found to facilitate HUVECs proliferation, migration and angiogenesis under hyperglycemia. In vivo experiments revealed JAM-A overexpression significantly enhanced MSCs survival, promoted wound angiogenesis, and thus accelerated diabetic wound closure, partially by enhancing PDGF-BB and VEGF expression. This study firstly demonstrated that JAM-A expression of MSCs was inhibited upon high-glucose stimulation. JAM-A overexpression alleviated high-glucose-induced MSCs dysfunction, enhanced their anti-oxidative capability, protected MSCs from hyperglycemia-induced apoptosis and improved their survival, thus strengthening MSCs paracrine function to promote angiogenesis and significantly accelerating diabetic wound healing, which offers a promising strategy to maximize MSCs-based therapy in diabetic wound.


Asunto(s)
Diabetes Mellitus , Hiperglucemia , Células Madre Mesenquimatosas , Neovascularización Fisiológica , Cicatrización de Heridas , Heridas y Lesiones , Humanos , Becaplermina/genética , Becaplermina/metabolismo , Supervivencia Celular/genética , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Glucosa/farmacología , Hiperglucemia/genética , Hiperglucemia/metabolismo , Células Madre Mesenquimatosas/metabolismo , Neovascularización Fisiológica/genética , Comunicación Paracrina/genética , Cordón Umbilical/citología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Cicatrización de Heridas/genética , Heridas y Lesiones/genética , Heridas y Lesiones/metabolismo
8.
Hypertension ; 80(4): 740-753, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36655574

RESUMEN

BACKGROUND: Vascular smooth muscle cells (SMCs) plasticity is a central mechanism in cardiovascular health and disease. We aimed at providing cellular phenotyping, epigenomic and proteomic depiction of SMCs derived from induced pluripotent stem cells and evaluating their potential as cellular models in the context of complex diseases. METHODS: Human induced pluripotent stem cell lines were differentiated using RepSox (R-SMCs) or PDGF-BB (platelet-derived growth factor-BB) and TGF-ß (transforming growth factor beta; TP-SMCs), during a 24-day long protocol. RNA-Seq and assay for transposase accessible chromatin-Seq were performed at 6 time points of differentiation, and mass spectrometry was used to quantify proteins. RESULTS: Both induced pluripotent stem cell differentiation protocols generated SMCs with positive expression of SMC markers. TP-SMCs exhibited greater proliferation capacity, migration and lower calcium release in response to contractile stimuli, compared with R-SMCs. Genes involved in the contractile function of arteries were highly expressed in R-SMCs compared with TP-SMCs or primary SMCs. R-SMCs and coronary artery transcriptomic profiles were highly similar, characterized by high expression of genes involved in blood pressure regulation and coronary artery disease. We identified FOXF1 and HAND1 as key drivers of RepSox specific program. Extracellular matrix content contained more proteins involved in wound repair in TP-SMCs and higher secretion of basal membrane constituents in R-SMCs. Open chromatin regions of R-SMCs and TP-SMCs were significantly enriched for variants associated with blood pressure and coronary artery disease. CONCLUSIONS: Both induced pluripotent stem cell-derived SMCs models present complementary cellular phenotypes of high relevance to SMC plasticity. These cellular models present high potential to study functional regulation at genetic risk loci of main arterial diseases.


Asunto(s)
Enfermedad de la Arteria Coronaria , Células Madre Pluripotentes Inducidas , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Transcriptoma , Proteómica , Enfermedad de la Arteria Coronaria/metabolismo , Diferenciación Celular/genética , Becaplermina/genética , Becaplermina/metabolismo , Becaplermina/farmacología , Factor de Crecimiento Transformador beta/metabolismo , Células Cultivadas , Miocitos del Músculo Liso/metabolismo , Cromatina/metabolismo
9.
Cell Mol Biol (Noisy-le-grand) ; 68(7): 123-128, 2022 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-36495508

RESUMEN

Vascular calcification is one of the major complications of chronic kidney disease (CKD), which could be further accelerated by the osteogenic transition and apoptosis of smooth muscle cells, thereby advancing the progression of renal diseases and increasing the mortality rate of cardiovascular events. MicroRNA is a kind of key regulator in the phenotypic transition of vascular smooth muscle cells (VSMCs), but its role remains unclear in VSMCs. In this study, VSMCs were stimulated by platelet-derived growth factors - BB (PDGF-BB) in varying concentrations to establish the VSMC dysfunction models. The relative expression of miR-29a-5p was quantified via the quantitative real-time polymerase chain reaction (qRT-PCR). The proliferation of VSMCs was determined via the BrdU method, analysis of cell cycle via flow cytometry, and the migration of VSMCs via Transwell assay. Expression of γ-secretase activating protein (GSAP) and markers of VSMC differentiation, including α-SMA, SM-22α, SMMHC and Calponin, was quantified via the Western blot. The targeting relationship between the 3'-UTR of miR-29a-5p and GSAP was validated through the dual-luciferase reporter gene assay. As a result, we found that PDGF-BB could trigger a decrease of miR-29a-5p in a time- and dose-dependent manner (P < 0.05). Overexpression of miR-29a-5p could curb the effect of PDGF-BB on the proliferation and migration of VSMCs while upregulating the expression of markers of differentiation (P < 0.05). In addition, the expression of GSAP was also affected by the negative regulation of miR-29a-5p, while the restoration of GSAP eliminated the effect of miR-29a-5p on the VSMCs partially (P < 0.05). Moreover, vascular calcification models were also established in the CKD rats, suggesting that the inhibition of GSAP could prevent PTH-induced vascular calcification in CKD rats. In conclusion, miR-29a-5p could inhibit the PDGF-BB-induced proliferation, migration and phenotypic transition of VSMCs via targeting GSAP. Thus, miR-29a-5p/GSAP might be a potential target for the treatment of vascular calcification.


Asunto(s)
MicroARNs , Insuficiencia Renal Crónica , Calcificación Vascular , Ratas , Animales , Músculo Liso Vascular , Becaplermina/genética , Becaplermina/metabolismo , Becaplermina/farmacología , Proliferación Celular , Movimiento Celular/genética , Miocitos del Músculo Liso/metabolismo , MicroARNs/metabolismo , Regiones no Traducidas 3' , Ciclo Celular , Calcificación Vascular/genética , Calcificación Vascular/metabolismo , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/metabolismo , Células Cultivadas
10.
PLoS One ; 17(10): e0275227, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36206226

RESUMEN

INTRODUCTION: Primary brain calcification (PBC) is a rare and intractable neurodegenerative disease. SLC20A2 and PDGFB are two major causative genes. As there is no effective treatment to avoid further progression or to prevent the onset of the disease, the patients may experience psychological distress. There is a qualitative study on the experiences of patients with primary brain calcification with SLC20A2 variants. However, the experiences of patients with PDGFB variants of the disease have not been explored. The purpose of this study is to identify the experiences of patients with PDGFB variants after diagnosis. MATERIALS AND METHODS: Semi-structured interviews were conducted once or twice a year for three years with five patients over the age of 21. The data were analyzed using inductive qualitative methods. RESULTS: Seven categories, 15 subcategories, and 129 codes were extracted. The seven categories are as follows: [Shock at hearing the term 'brain calcification' for the first time], [Anxiety regarding the risk of heredity], [Anxiety, along with severe headaches, and various other symptoms], [Gratitude for the family members who care], [Accepting the disease as a non-life-threatening illness], [Feeling alienated due to the rare intractable disease], and [Modifying lifestyle due to the illness]. DISCUSSION: The most stressful aspect of the disease was the headache that persisted even with the use of analgesics, which was different from patients with the SLC20A2 variants. In addition, we found unique concepts such as anxiety regarding the risk of heredity and a feeling of alienation due to the rare and intractable disease.


Asunto(s)
Encefalopatías , Calcinosis , Enfermedades Neurodegenerativas , Becaplermina/genética , Encéfalo/metabolismo , Encefalopatías/diagnóstico , Calcinosis/diagnóstico , Calcinosis/genética , Humanos , Mutación , Enfermedades Neurodegenerativas/genética , Proteínas Proto-Oncogénicas c-sis/genética , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/genética , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/metabolismo
11.
J Clin Lab Anal ; 36(10): e24683, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36059119

RESUMEN

OBJECT: The aim of the present work was to investigate the correlation of plasma platelet-derived growth factor (PDGF)-BB level and single nucleotide polymorphism (SNP, rs1800817 and rs2285094) of PDGF-B gene with the onset and stability condition of coronary heart disease (CHD). METHODS: Totally, 335 subjects were included in and divided into CHD (n = 247) and control group (n = 88) according to coronary angiography. Besides, the patients in the CHD group were divided into acute coronary syndrome (ACS) group (n = 165) and stable angina pectoria (SAP) group (n = 82), based on CHD stability condition. The plasma PDGF-BB level was measured by ELISA, and the genotype of PDGF-B was examined through qPCR assay. RESULTS: The PDGF-BB level was positively correlated with hsCRP level (r = 0.149, p < 0.05). The genotype frequencies of SNP rs1800817 and rs2285094 match Hardy-Weinberg equilibrium. There was weak linkage disequilibrium between SNP rs1800817 and rs2285094: D' = 0.419, r2  = 0.04, which has no correlation with CHD. There was no statistical difference in plasma PDGF-BB level among different genotypes in rs1800817 and rs2285094. There were no differences in the plasma PDGF-BB level among patients with any genotype of SNP rs1800817 and rs2285094, no matter how it was grouped. Logistic regression results indicated that the plasma PDGF-BB level was the independent risk factor of CHD onset (OR = 1.003, 95% CI 1.001-1.006, p = 0.014). CONCLUSIONS: High plasma PDGF-BB level is the risk factor of CHD and has correlation with instability of CHD. The plasma PDGF-BB level change may be related to inflammatory response. PDGF-B gene rs1800817 and rs2285094 polymorphisms are not correlated with CHD.


Asunto(s)
Enfermedad Coronaria , Infarto del Miocardio , Becaplermina/genética , Proteína C-Reactiva , Enfermedad Coronaria/epidemiología , Enfermedad Coronaria/genética , Humanos , Polimorfismo de Nucleótido Simple/genética
12.
Sci Rep ; 12(1): 10261, 2022 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-35715541

RESUMEN

The A1762T/G1764A double mutant in the basal core promoter (BCP) region of the hepatitis B virus (HBV) is associated with severe hepatic lesions while the G1899A mutation with the double mutant is associated with a significant reduction in the risk of severe fibrosis. This study aims to measure a number of markers in the serum of patients with chronic HBV infection and to assess relationships between these markers and BCP/precore mutants with consideration of the stage of fibrosis. The serum levels of resistin, TGF-ß1, MMP-1, TIMP-1, collagen IA1 and PDGF-BB, which are markers that are known to be involved in the process of hepatic fibrosis, were assayed. The serum levels of PDGF-BB and TIMP-1, and the mutation profile were independently associated with advanced fibrosis. A higher level of TIMP-1 was associated with advanced fibrosis regardless of the mutation status, and a higher level of PDGF-BB was associated with nonsevere fibrosis in patients infected with viruses harboring the A1762T/G1764A or A1762T/G1764A/G1899A mutations. Our results suggest an impact of the A1762T/G1764A mutant on the biological pathway related to TGF-ß1 and PDGF-BB. In vitro studies are needed to understand the impact of these mutants on the serum secretion of markers involved in fibrosis severity.


Asunto(s)
Hepatitis B Crónica , Hepatitis B , Becaplermina/genética , Biomarcadores , ADN Viral/genética , Genotipo , Virus de la Hepatitis B/genética , Hepatitis B Crónica/complicaciones , Humanos , Cirrosis Hepática/complicaciones , Mutación , Inhibidor Tisular de Metaloproteinasa-1/genética , Factor de Crecimiento Transformador beta1/genética
13.
Biochem Biophys Res Commun ; 593: 93-100, 2022 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-35063775

RESUMEN

Inorganic phosphate (Pi) is the second most abundant inorganic ion in the body. Since abnormalities in Pi metabolism are risk factors for various diseases, serum Pi levels are strictly controlled. Type-III sodium-dependent Pi transporters, PiT-1 (encoded by SLC20A1) and PiT-2 (encoded by SLC20A2), are distributed throughout the tissues of the body, including the central nervous system, and are known to be responsible for extracellular to intracellular Pi transport. Platelet-derived growth factor (PDGF) is a major growth factor of mesenchymal cells. PDGF-BB, a homodimer of PDGF-B, regulates intracellular Pi by increasing PiT-1 expression in vascular smooth muscle cells. However, the effects of PDGF-BB on Pi transporters in neurons have yet to be reported. Here, we investigated the effect of PDGF-BB on Pi transporters in human neuroblastoma SH-SY5Y cells. PDGF-BB did not induce SLC20A1 mRNA expression, but it increased the intracellular uptake of Pi via PiT-1 in SH-SY5Y cells. Among the signaling pathways associated with PDGF-BB, AKT signaling was shown to be involved in the increase in Pi transport. In addition, the PDGF-BB-induced increase in Pi mediated neuroprotective effects in SLC20A2-suppressed cells, in an in vitro model of the pathological condition found in idiopathic basal ganglia calcification. Moreover, the increase in Pi uptake was found to occur through promotion of intracellular PiT-1 translocation to the plasma membrane. Overall, these results indicate that PDGF-BB exerts neuroprotective effects via Pi transport, and they demonstrate the potential utility of PDGF-BB against abnormal Pi metabolism in neurons.


Asunto(s)
Becaplermina/metabolismo , Neuroblastoma/metabolismo , Fosfatos/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/metabolismo , Becaplermina/genética , Transporte Biológico , Humanos , Neuroblastoma/genética , Neuroblastoma/patología , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/genética , Células Tumorales Cultivadas
14.
Genes Genomics ; 44(4): 395-404, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35066809

RESUMEN

BACKGROUND: We have clarified the role of miR-382-3p in chronic thromboembolic pulmonary hypertension (CTEPH), but what is less clear lies in its upstream regulatory mechanism. OBJECTIVE: To explore the regulation mechanism of GAS5/miR-382-3p axis on CTEPH. METHODS: In vitro, we constructed cell models by treating Pulmonary Artery Smooth Muscle Cells (PASMCs) with platelet-derived growth factor-BB (PDGF-BB). The effects of different concentrations of PDGF-BB on the activity of PASMCs were tested by cell counting kit-8 (CCK-8). The upstream lncRNA of miR-382-3p was screened and confirmed through bioinformatics analysis, RNA pull-down, quantitative reverse transcription polymerase chain reaction (qRT-PCR), dual luciferase reporter gene and RNA immunoprecipitation assays. The effects of GAS5/miR-382-3p axis on the viability, migration, and expressions of autophagy- and angiogenesis-related proteins were confirmed by rescue experiments (CCK-8, wound healing and western blot). In vivo, animal models by perfusing autologous blood vessels, the effects of GAS5 overexpression or silencing on the expressions of miR-382-3p, angiogenesis- and autophagy-related genes, mean pulmonary arterial pressure (mPAP) and pulmonary artery wall were determined by biological signal acquisition system, hematoxylin-eosin staining, qRT-PCR and western blot. RESULTS: PDGF-BB dose-dependently promoted PASMCs viability. XIST and GAS5 expressions in PASMCs were affected by the concentration of PDGF-BB, but only GAS5 can be pulled down by miR-382-3p probe. GAS5 targeted miR-382-3p to inhibit the viability and migration of PAMSCs, mPAP in CTEPH rats, pulmonary artery wall thickening and angiogenesis, and promote autophagy. CONCLUSIONS: GAS5/miR-382-3p axis is involved in the regulation of pulmonary artery remodeling and autophagy in CTEPH.


Asunto(s)
Hipertensión Pulmonar , MicroARNs , Arteria Pulmonar , ARN Largo no Codificante , Animales , Autofagia/genética , Becaplermina/genética , Becaplermina/farmacología , Humanos , Hipertensión Pulmonar/genética , MicroARNs/genética , Arteria Pulmonar/metabolismo , ARN Largo no Codificante/genética , Ratas
15.
Sci Rep ; 12(1): 795, 2022 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-35039587

RESUMEN

The epidermal basement membrane deteriorates with aging. We previously reported that basement membrane reconstruction not only serves to maintain epidermal stem/progenitor cells in the epidermis, but also increases collagen fibrils in the papillary dermis. Here, we investigated the mechanism of the latter action. Collagen fibrils in the papillary dermis were increased in organotypic human skin culture treated with matrix metalloproteinase and heparinase inhibitors. The expression levels of COL5A1 and COL1A1 genes (encoding collagen type V α 1 chain and collagen type I α 1 chain, respectively) were increased in fibroblasts cultured with conditioned medium from a skin equivalent model cultured with the inhibitors and in keratinocytes cultured on laminin-511 E8 fragment-coated plates. We then examined cytokine expression, and found that the inhibitors increased the expression of PDGF-BB (platelet-derived growth factor consisting of two B subunits) in epidermis. Expression of COL5A1 and COL1A1 genes was increased in cultured fibroblasts stimulated with PDGF-BB. Further, the bifunctional inhibitor hydroxyethyl imidazolidinone (HEI) increased skin elasticity and the thickness of the papillary dermis in the skin equivalent. Taken together, our data suggests that reconstructing the basement membrane promotes secretion of PDGF-BB by epidermal keratinocytes, leading to increased collagen expression at the papillary dermis.


Asunto(s)
Membrana Basal/fisiología , Epidermis/fisiología , Colágenos Asociados a Fibrillas/fisiología , Fibroblastos/metabolismo , Fibroblastos/fisiología , Regeneración/fisiología , Envejecimiento de la Piel/patología , Envejecimiento de la Piel/fisiología , Membrana Basal/metabolismo , Becaplermina/genética , Becaplermina/metabolismo , Células Cultivadas , Cadena alfa 1 del Colágeno Tipo I/genética , Cadena alfa 1 del Colágeno Tipo I/metabolismo , Colágeno Tipo V/genética , Colágeno Tipo V/metabolismo , Células Epidérmicas/metabolismo , Epidermis/metabolismo , Epidermis/patología , Colágenos Asociados a Fibrillas/genética , Colágenos Asociados a Fibrillas/metabolismo , Expresión Génica , Humanos , Queratinocitos/metabolismo , Metaloproteinasas de la Matriz/farmacología , Regeneración/genética
16.
Biochim Biophys Acta Mol Basis Dis ; 1868(1): 166278, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34601016

RESUMEN

BACKGROUND: The vascular pathology of peripheral artery disease (PAD) encompasses abnormal microvascular architecture and fibrosis in response to ischemia-reperfusion (I/R) cycles. We aimed to investigate the mechanisms by which pathological changes in the microvasculature direct fibrosis in the context of I/R. METHODS: Primary human aortic endothelial cells (ECs) were cultured under cycles of normoxia-hypoxia (NH) or normoxia-hypoxia-hyperoxia (NHH) to mimic I/R. Primary human aortic smooth muscle cells (SMCs) were cultured and treated with media from the ECs. FINDINGS: The mRNA and protein expression of the pro-fibrotic factors platelet derived growth factor (PDGF)-BB and connective tissue growth factor (CTGF) were significantly upregulated in ECs undergoing NH or NHH cycles. Treatment of SMCs with media from ECs undergoing NH or NHH cycles led to significant increases in TGF-ß1, TGF-ß pathway signaling intermediates, and collagen expression. Addition of neutralizing antibodies against PDGF-BB and CTGF to the media blunted the increases in TGF-ß1 and collagen expression. Treatment of SMCs with PAD patient-derived serum also led to increased TGF-ß1 levels. INTERPRETATION: In an in-vitro model of I/R, which recapitulates the pathophysiology of PAD, increased secretion of PDGF-BB and CTGF by ECs was shown to be predominantly driving TGF-ß1-mediated expression by SMCs. These cell culture experiments help elucidate the mechanism and interaction between ECs and SMCs in microvascular fibrosis associated with I/R. Thus, targeting these pro-fibrotic factors may be an effective strategy to combat fibrosis in response to cycles of I/R. FUNDING: National Institute on Aging at the National Institutes of Health grant number R01AG064420. RESEARCH IN CONTEXT: Evidence before this study: Previous studies in gastrocnemius biopsies from peripheral artery disease (PAD) patients showed that transforming growth factor beta 1 (TGF-ß1), the most potent inducer of pathological fibrosis, is increased in the vasculature of PAD patients and correlated with collagen deposition. However, the exact cellular source of TGF-ß1 remained unclear. Added value of this study: Exposing cells to cycles of normoxia-hypoxia-hyperoxia (NHH) resulted in pathological changes that are consistent with human PAD. This supports the idea that the use of NHH may be a reliable, novel in vitro model of PAD useful for studying associated pathophysiological mechanisms. Furthermore, pro-fibrotic factors (PDGF-BB and CTGF) released from endothelial cells were shown to induce a fibrotic phenotype in smooth muscle cells. This suggests a potential interaction between these cell types in the microvasculature that drives increased TGF-ß1 expression and collagen deposition. Thus, targeting these pro-fibrotic factors may be an effective strategy to combat fibrosis in response to cycles of ischemia-reperfusion.


Asunto(s)
Becaplermina/genética , Factor de Crecimiento del Tejido Conjuntivo/genética , Enfermedad Arterial Periférica/genética , Factor de Crecimiento Transformador beta1/genética , Aorta/metabolismo , Aorta/patología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Fibrosis/genética , Fibrosis/patología , Regulación de la Expresión Génica/genética , Humanos , Hiperoxia/genética , Hiperoxia/patología , Hipoxia/genética , Hipoxia/patología , Microvasos/metabolismo , Microvasos/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Enfermedad Arterial Periférica/patología , Cultivo Primario de Células , Transducción de Señal/genética
17.
Nat Commun ; 12(1): 6219, 2021 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-34711819

RESUMEN

Vascularization is critical for skull development, maintenance, and healing. Yet, there remains a significant knowledge gap in the relationship of blood vessels to cranial skeletal progenitors during these processes. Here, we introduce a quantitative 3D imaging platform to enable the visualization and analysis of high-resolution data sets (>100 GB) throughout the entire murine calvarium. Using this technique, we provide single-cell resolution 3D maps of vessel phenotypes and skeletal progenitors in the frontoparietal cranial bones. Through these high-resolution data sets, we demonstrate that CD31hiEmcnhi vessels are spatially correlated with both Osterix+ and Gli1+ skeletal progenitors during postnatal growth, healing, and stimulated remodeling, and are concentrated at transcortical canals and osteogenic fronts. Interestingly, we find that this relationship is weakened in mice with a conditional knockout of PDGF-BB in TRAP+ osteoclasts, suggesting a potential role for osteoclasts in maintaining the native cranial microvascular environment. Our findings provide a foundational framework for understanding how blood vessels and skeletal progenitors spatially interact in cranial bone, and will enable more targeted studies into the mechanisms of skull disease pathologies and treatments. Additionally, our technique can be readily adapted to study numerous cell types and investigate other elusive phenomena in cranial bone biology.


Asunto(s)
Neovascularización Fisiológica , Cráneo/irrigación sanguínea , Animales , Becaplermina/genética , Becaplermina/metabolismo , Imagenología Tridimensional , Ratones , Ratones Endogámicos C57BL , Microcirculación , Osteoclastos/metabolismo , Cráneo/diagnóstico por imagen , Cráneo/metabolismo
18.
Bioengineered ; 12(2): 9463-9472, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34699319

RESUMEN

Vascular smooth muscle cell (VSMC) hyperplasia is closely associated with AS progression. Hence, it is of great significance to elucidate the molecular mechanisms underlying the involvement of VSMCs in AS. SHH antagonist can inhibit the excessive proliferation, migration and phenotypic transformation of PDGF-BB-induced VSMCs. It has been proved that CUL3 can suppress Hedgehog signaling. This current work was designed to identify the biological role of CUL3 in the behaviors of VSMCs in AS and investigate the potential molecular mechanism. VSMCs were treated with PDGF-BB to establish the cell model in vitro. Levels of CUL3, SHH and Gli1 in PDGF-BB-stimulated VSMCs were measured by RT-qPCR analysis. Then, the precise functions of CUL3 in VSMCs were determined from the perspectives of proliferation, migration, apoptosis and phenotype transformation. Besides, the influence of CUL3 on inflammatory response in VSMCs was evaluated. Moreover, the impact of CUL3 on Hedgehog signaling pathway was also investigated. In the present research, it was observed that CUL3 was lowly expressed and SHH and Gli1 were highly expressed in PDGF-BB-stimulated VSMCs. Upregulation of CUL3 suppressed the excessive proliferation, migration and phenotypic transformation and facilitated the apoptosis of PDGF-BB-stimulated VSMCs. In addition, elevation of CUL3 alleviated inflammatory response in PDGF-BB-stimulated VSMCs. Importantly, CUL3 overexpression inactivated Hedgehog signaling pathway. To conclude, CUL3 might regulate the biological behaviors of VSMCs in AS by modulating Hedgehog signaling pathway. These data encourage to further investigate any potential therapeutic role of CUL3 in animal models of AS and explore therapeutic values for AS clinically.


Asunto(s)
Becaplermina/metabolismo , Movimiento Celular , Proliferación Celular , Proteínas Cullin/metabolismo , Proteínas Hedgehog/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Transducción de Señal , Becaplermina/genética , Proteínas Cullin/genética , Proteínas Hedgehog/genética , Humanos
19.
Nanotechnology ; 32(44)2021 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-34298537

RESUMEN

An innovative surface-enhanced Raman spectroscopy and lateral flow assay (SERS-LFA) biosensor combined with aptamer recognition had been developed for the convenient, rapid, sensitive and accurate detection of thrombin and platelet-derived growth factor-BB (PDGF-BB) associated with prostate cancer simultaneously. During the biosensor operation, thrombin and PDGF-BB in the sample were recognized and combined by thiol-modified aptamers immobilized on Au-Ag hollow nanoparticles (Au-Ag HNPs) surface and biotinylated aptamers immobilized on the test lines of the biosensor. Thus, thrombin and PDGF-BB were simultaneously captured between detection aptamers and capture aptamers in a sandwich structure. Finite difference time domain simulation confirmed that 'hot spots' appeared at the gaps of Au-Ag HNPs dimer in the enhanced electromagnetic field compared to that of a single Au-Ag HNP, indicating that the aggregated Au-Ag HNPs owned a good SERS signal amplification effect. The detection limits of thrombin and PDGF-BB in human plasma were as low as 4.837 pg ml-1and 3.802 pg ml-1, respectively. Moreover, the accuracy of the biosensor which was applied to detect thrombin and PDGF-BB in prostate cancer plasma had been verified. This designed biosensor had broad application prospects in the clinical diagnosis of prostate cancer.


Asunto(s)
Becaplermina/sangre , Técnicas Biosensibles/métodos , Neoplasias de la Próstata/sangre , Espectrometría Raman/métodos , Trombina/análisis , Anciano , Anticuerpos Monoclonales , Aptámeros de Nucleótidos , Becaplermina/genética , Técnicas Biosensibles/instrumentación , Análisis Químico de la Sangre/métodos , Oro/química , Humanos , Límite de Detección , Masculino , Nanopartículas del Metal/química , Persona de Mediana Edad , Oxazinas/química , Sensibilidad y Especificidad , Plata/química , Trombina/genética
20.
Int J Mol Sci ; 22(8)2021 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-33919968

RESUMEN

The aim of the present study was to investigate the influence of a novel volume-stable collagen matrix (vCM) on early wound healing events including cellular migration and adhesion, protein adsorption and release, and the dynamics of the hemostatic system. For this purpose, we utilized transwell migration and crystal violet adhesion assays, ELISAs for quantification of adsorbed and released from the matrix growth factors, and qRT-PCR for quantification of gene expression in cells grown on the matrix. Our results demonstrated that primary human oral fibroblasts, periodontal ligament, and endothelial cells exhibited increased migration toward vCM compared to control cells that migrated in the absence of the matrix. Cellular adhesive properties on vCM were significantly increased compared to controls. Growth factors TGF-ß1, PDGF-BB, FGF-2, and GDF-5 were adsorbed on vCM with great efficiency and continuously delivered in the medium after an initial burst release within hours. We observed statistically significant upregulation of genes encoding the antifibrinolytic thrombomodulin, plasminogen activator inhibitor type 1, thrombospondin 1, and thromboplastin, as well as strong downregulation of genes encoding the profibrinolytic tissue plasminogen activator, urokinase-type plasminogen activator, its receptor, and the matrix metalloproteinase 14 in cells grown on vCM. As a general trend, the stimulatory effect of the vCM on the expression of antifibrinolytic genes was synergistically enhanced by TGF-ß1, PDGF-BB, or FGF-2, whereas the strong inhibitory effect of the vCM on the expression of profibrinolytic genes was reversed by PDGF-BB, FGF-2, or GDF-5. Taken together, our data strongly support the effect of the novel vCM on fibrin clot stabilization and coagulation/fibrinolysis equilibrium, thus facilitating progression to the next stages of the soft tissue healing process.


Asunto(s)
Colágeno/farmacología , Mucosa Bucal/efectos de los fármacos , Ligamento Periodontal/efectos de los fármacos , Regeneración/genética , Cicatrización de Heridas/genética , Animales , Becaplermina/genética , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Colágeno/química , Células Endoteliales/efectos de los fármacos , Fibrina/genética , Fibrinólisis/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/genética , Fibroblastos/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Factor 5 de Diferenciación de Crecimiento/genética , Hemostasis/efectos de los fármacos , Xenoinjertos , Humanos , Ratones , Mucosa Bucal/crecimiento & desarrollo , Ligamento Periodontal/crecimiento & desarrollo , Cultivo Primario de Células , Factor de Crecimiento Transformador beta1/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA