Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
2.
J Cancer Res Clin Oncol ; 150(4): 214, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38662050

RESUMO

PURPOSE: Core 1ß1,3-galactosyltransferase 1 (C1GALT1) exhibits elevated expression in multiple cancers. The present study aimed to elucidate the clinical significance of C1GALT1 aberrant expression and its impact on radiosensitivity in lung adenocarcinoma (LUAD). METHODS: The C1GALT1 expression and its clinical relevance were investigated through public databases and LUAD tissue microarray analyses. A549 and H1299 cells with either C1GALT1 knockdown or overexpression were further assessed through colony formation, gamma-H2A histone family member X immunofluorescence, 5-ethynyl-2'-deoxyuridine incorporation, and flow cytometry assays. Bioinformatics analysis was used to explore single cell sequencing data, revealing the influence of C1GALT1 on cancer-associated cellular states. Vimentin, N-cadherin, and E-cadherin protein levels were measured through western blotting. RESULTS: The expression of C1GALT1 was significantly higher in LUAD tissues than in adjacent non-tumor tissues both at mRNA and protein level. High expression of C1GALT1 was correlated with lymph node metastasis, advanced T stage, and poor survival, and was an independent risk factor for overall survival. Radiation notably upregulated C1GALT1 expression in A549 and H1299 cells, while radiosensitivity was increased following C1GALT1 knockdown and decreased following overexpression. Experiment results showed that overexpression of C1GALT1 conferred radioresistance, promoting DNA repair, cell proliferation, and G2/M phase arrest, while inhibiting apoptosis and decreasing E-cadherin expression, alongside upregulating vimentin and N-cadherin in A549 and H1299 cells. Conversely, C1GALT1 knockdown had opposing effects. CONCLUSION: Elevated C1GALT1 expression in LUAD is associated with an unfavorable prognosis and contributes to increased radioresistance potentially by affecting DNA repair, cell proliferation, cell cycle regulation, and epithelial-mesenchymal transition (EMT).


Assuntos
Adenocarcinoma de Pulmão , Galactosiltransferases , Neoplasias Pulmonares , Tolerância a Radiação , Humanos , Adenocarcinoma de Pulmão/patologia , Adenocarcinoma de Pulmão/genética , Adenocarcinoma de Pulmão/metabolismo , Adenocarcinoma de Pulmão/radioterapia , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Transição Epitelial-Mesenquimal , Galactosiltransferases/genética , Galactosiltransferases/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/radioterapia , Neoplasias Pulmonares/mortalidade , Prognóstico , Tolerância a Radiação/genética
3.
Tissue Eng Part B Rev ; 30(2): 198-216, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37658839

RESUMO

The development of tracheal tissue engineering (TTE) has seen a rapid growth in recent years. The purpose of this study was to investigate the global status, trends, and hotspots of TTE research based on bibliometrics and visualization analysis. Publications related to TTE were retrieved and included in the Web of Science Core Collection. VOSviewer and CiteSpace were used to generate knowledge maps. Six hundred fifty-five publications were identified, and the quantity of the annual publications worldwide was on the increase. International collaboration is a widespread reality. The United States led the world in the field of trachea tissue engineering, whereas University College London was the institution with the greatest contribution. In addition, Biomaterials had a great influence in this field, attracting the largest number of papers. Moreover, the topics of TTE research largely concentrated on the biomechanical scaffold preparation, the vascularization and epithelialization of scaffold, the tracheal cartilage regeneration, and the tissue-engineered tracheal transplantation. And the research on the application of decellularization and 3D printing for the construction of a tissue-engineered trachea was likely to receive more widespread attention in the future. Impact statement In recent years, tracheal tissue engineering (TTE) has experienced rapid growth. In this study, we investigated the worldwide status and trends of TTE research, and revealed the countries, institutions, journals, and authors that had made significant contributions to the field of TTE. Moreover, the possible research hotspots in the future were predicted. According to our research, researchers can gain a better understanding of the trends in this field, and stay informed of the most current research by tracking key journals, institutions, and authors.


Assuntos
Engenharia Tecidual , Traqueia , Humanos , Bibliometria , Materiais Biocompatíveis , Impressão Tridimensional
4.
Biotechnol J ; 19(1): e2300040, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37985427

RESUMO

In this study, we aimed to utilize autologous tracheal epithelia and BMSCs as the seeding cells, utilize PCL coated with SilMA as the hybrid scaffold to carry the cells and KGN, which can selectively stimulate chondrogenic differentiation of BMSCs. This hybrid tracheal substitution was carried out to repair the tracheal partial window-shape defect. Firstly, SilMA with the concentration of 10%, 15% and 20% was prepared, and the experiment of swelling and degradation was performed. With the increase of the concentration, the swelling ratio of SilMA decreased, and the degradation progress slowed down. Upon the result of CCK-8 test and HE staining of 3D co-culture, the SilMA with concentration of 20% was selected. Next, SilMA and the cells attached to SilMA were characterized by SEM. Furthermore, in vitro cytotoxicity test shows that 20% SilMA has good cytocompatibility. The hybrid scaffold was then made by PCL coated with 20% SilMA. The mechanical test shows this hybrid scaffold has better biomechanical properties than native trachea. In vivo tracheal defect repair assays were conducted to evaluate the effect of the hybrid substitution. H&E staining, IHC staining and IF staining showed that this hybrid substitution ensured the viability, proliferation and migration of epithelium. However, it is sad that the results of chondrogenesis were not obvious. This study is expected to provide new strategies for the fields of tracheal replacement therapy needing mechanical properties and epithelization.


Assuntos
Fibroínas , Traqueia/cirurgia , Engenharia Tecidual/métodos , Técnicas de Cocultura , Impressão Tridimensional , Alicerces Teciduais
5.
Mater Today Bio ; 23: 100841, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37920292

RESUMO

Functional vascularization is crucial for maintaining the long-term patency of tissue-engineered trachea and repairing defective trachea. Herein, we report the construction and evaluation of a novel cell-free tissue-engineered tracheal scaffold that effectively promotes vascularization of the graft. Our findings demonstrated that exosomes derived from endothelial progenitor cells (EPC-Exos) enhance the proliferation, migration, and tube formation of endothelial cells. Taking advantage of the angiogenic properties of EPC-Exos, we utilized methacrylate gelatin (GelMA) as a carrier for endothelial progenitor cell exosomes and encapsulated them within a 3D-printed polycaprolactone (PCL) scaffold to fabricate a composite tracheal scaffold. The results demonstrated the excellent angiogenic potential of the methacrylate gelatin/vascular endothelial progenitor cell exosome/polycaprolactone tracheal scaffold. Furthermore, in vivo reconstruction of tracheal defects revealed the capacity of this composite tracheal stent to remodel vasculature. In conclusion, we have successfully developed a novel tracheal stent composed of methacrylate gelatin/vascular endothelial progenitor exosome/polycaprolactone, which effectively promotes angiogenesis for tracheal repair, thereby offering significant prospects for clinical and translational medicine.

6.
Bioengineered ; 14(1): 2274150, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37927226

RESUMO

Transplantation of tissue-engineered trachea is an effective treatment for long-segment tracheal injury. This technology avoids problems associated with a lack of donor resources and immune rejection, generating an artificial trachea with good biocompatibility. To our knowledge, a systematic summary of basic and clinical research on tissue-engineered trachea in the last 20 years has not been conducted. Here, we analyzed the development trends of tissue-engineered trachea research by bibliometric means and outlined the future perspectives in this field. The Web of Science portal was selected as the data source. CiteSpace, VOSviewer, and the Bibliometric Online Analysis Platform were used to analyze the number of publications, journals, countries, institutions, authors, and keywords from 475 screened studies. Between 2000 and 2023, the number of published studies on tissue-engineered trachea has been increasing. Biomaterials published the largest number of papers. The United States and China have made the largest contributions to this field. University College London published the highest number of studies, and the most productive researcher was an Italian scholar, Paolo Macchiarini. However, close collaborations between various researchers and institutions from different countries were generally lacking. Despite this, keyword analysis showed that manufacturing methods for tracheal stents, hydrogel materials, and 3D bioprinting technology are current popular research topics. Our bibliometric study will help scientists in this field gain an in-depth understanding of the current research progress and development trends to guide their future work, and researchers in related fields will benefit from the introduction to transplantation methods of tissue-engineered trachea.


We conducted a comprehensive bibliometric analysis of tissue-engineered trachea.We systematically outlined the preparation methods and current development forms of tissue-engineered trachea.We predicted future tissue-engineered trachea research trends from the perspectives of countries, institutions, researchers, and popular research topics.


Assuntos
Engenharia Tecidual , Traqueia , Humanos , Traqueia/cirurgia , Bibliometria , Materiais Biocompatíveis , China
7.
Bioeng Transl Med ; 8(4): e10534, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37476057

RESUMO

Formation of functional and perfusable vascular network is critical to ensure the long-term survival and functionality of the engineered tissue tracheae after transplantation. However, the greatest challenge in tracheal-replacement therapy is the promotion of tissue regeneration by rapid graft vascularization. Traditional prevascularization methods for tracheal grafts typically utilize omentum or muscle flap wrapping, which requires a second operation; vascularized segment tracheal orthotopic transplantation in one step remains difficult. This study proposes a method to construct a tissue-engineered tracheal graft, which directly forms the microvascular network after orthotopic transplantation in vivo. The focus of this study was the preparation of a hybrid tracheal graft that is non-immunogenic, has good biomechanical properties, supports cell proliferation, and quickly vascularizes. The results showed that vacuum-assisted decellularized trachea-polycaprolactone hybrid scaffold could match most of the above requirements as closely as possible. Furthermore, endothelial progenitor cells (EPCs) were extracted and used as vascularized seed cells and seeded on the surfaces of hybrid grafts before and during the tracheal orthotopic transplantation. The results showed that the microvascularized tracheal grafts formed maintained the survival of the recipient, showing a satisfactory therapeutic outcome. This is the first study to utilize EPCs for microvascular construction of long-segment trachea in one-step; the approach represents a promising method for microvascular tracheal reconstruction.

9.
Mol Med Rep ; 27(5)2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36960863

RESUMO

Subsequently to the publication of the above paper, an interested reader drew to the authors' attention that, for the Transwell invasion assay experiments with the SK­MES­1 cell line shown in Fig. 4A on p. 1748, the 'mimic'NC' and 'inhibitor­NC' data panels showed overlapping sections, such that these data may have been derived from the same original source even though they were intending to show the results of different experiments. The authors have consulted their original data, and realize that the 'inhibitor­NC' data panel was inadvertently selected incorrectly for Fig. 4A. The revised version of Fig. 4, showing the correct data for the 'inhibitor­NC' experiment, is shown on the next page. Note that the error made during the assembly of Fig. 4 did not significantly affect either the results or the conclusions reported in this paper, and all the authors agree to this Corrigendum. The authors are grateful to the Editor of Molecular Medicine Reports for allowing them the opportunity to publish this corrigendum, and apologize to the readership for any inconvenience caused. [Molecular Medicine Reports 17: 1742­1752, 2018; DOI: 10.3892/mmr.2017.8050].

10.
Chem Biol Interact ; 368: 110248, 2022 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-36343684

RESUMO

UGT1A1 is the main enzyme that catalyzes the metabolic elimination and detoxification of SN-38, the active form of the drug irinotecan. Milk thistle products have been used widely to protect the liver from injury associated with the use of chemotherapeutic agents. To evaluate whether SN-38 metabolism can be affected by milk thistle products, the inhibitory effects of silybins on UGT1A1*1 and UGT1A1*6 were evaluated in the present investigation. Both silybin A and silybin B potently inhibited SN-38 glucuronidation catalyzed by UGT1A1*1 or UGT1A1*6. It was noteworthy that silybin A and silybin B showed synergistic effect in UGT1A1*1 microsomes at concentration around IC50, while additive effect in UGT1A1*6. According to the predicted AUCi/AUC ratios (the ratio of the area under the plasma concentration-time curve of SN-38 in the presence and absence of silybins), the coadministration of irinotecan and several milk thistle products, including silybin-phosphatidylcholine complex, two Legalon capsules, four Silymarin tablets or four Liverman capsules, may lead to clinically significant herb-drug interactions (HDI) via UGT1A1 inhibition. Meanwhile, Rgut values were much higher than 11 in all the groups, indicating potential HDI due to intestinal UGT1A1 inhibition.


Assuntos
Glucuronosiltransferase , Silybum marianum , Irinotecano/metabolismo , Silibina/metabolismo , Silibina/farmacologia , Glucuronosiltransferase/metabolismo , Microssomos Hepáticos/metabolismo , Catálise , Camptotecina
11.
ACS Appl Mater Interfaces ; 14(38): 42791-42803, 2022 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-36112832

RESUMO

Fe3O4 nanoparticles are the most widely used magnetic nanoparticles in the biomedicine field. The biodistribution of most nanoparticles in vivo is determined by the capture of macrophages; however, the effects of nanoparticles on macrophages remain poorly understood. Here, we demonstrated that Fe3O4 nanoparticles could reduce macrophage viability after 48 h of treatment and induce a shift in macrophage polarization toward the M1 phenotype; RNA sequencing revealed the activation of the ferroptosis pathway and p53 upregulation compared to the control group. The expression in p53, xCT, glutathione peroxidase 4 (GPX4), and transferrin receptor (TFR) in macrophages was similar to that in erastin-induced ferroptosis in macrophages, and the ultrastructural morphology of mitochondria was consistent with that of erastin-treated cells. We used DCFH-DA to estimate the intracellular reactive oxygen species content in Fe3O4 nanoparticles treated with Ana-1 and JC-1 fluorescent probes to detect the mitochondrial membrane potential change; both showed to be time-dependent. Fer-1 inhibited the reduction of the glutathione/oxidized glutathione (GSH/GSSG) ratio and inhibited intracellular oxidative stress states; therefore, Fe3O4 nanoparticles induced ferroptosis in macrophages. Finally, we used pifithrin-α hydrobromide (PFT) as a p53 inhibitor to verify whether the high expression of p53 is involved in mediating this process. After PFT treatment, the live/dead cell rate, TFR, p53 expression, and GPX4 consumption were inhibited and mitigated the GSH/GSSG ratio reduction as well. This indicates that p53 may contribute to Fe3O4 nanoparticle-induced ferroptosis of macrophages. We provide a theoretical basis for the molecular mechanisms of ferroptosis in macrophages and the biotoxicity in vivo induced by Fe3O4 nanoparticles.


Assuntos
Ferroptose , Nanopartículas , Corantes Fluorescentes , Glutationa/metabolismo , Dissulfeto de Glutationa/metabolismo , Macrófagos/metabolismo , Fosfolipídeo Hidroperóxido Glutationa Peroxidase , Espécies Reativas de Oxigênio/metabolismo , Receptores da Transferrina/metabolismo , Distribuição Tecidual , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
12.
Ann Thorac Med ; 17(3): 173-179, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35968400

RESUMO

OBJECTIVES: To explore the clinical value of three-dimensional (3D) reconstruction technology combined with 3D printing in the treatment of pectus excavatum (PE). METHODS: The clinical data of 10 patients with PE in our department from June 2018 to December 2020 were analyzed retrospectively. All patients underwent thin-layer computed tomography examination before the operation, and then 3D reconstruction was performed with Mimics 20.0 software. The radian and curvature of the pectus bar were designed according to the reconstructed images. Afterward, the images were imported into the light-curing 3D printer in STL format for slice printing. Hence that the personalized operation scheme, including the size of the pectus bar and the surgical approach, can be made according to the 3D printed model. The thoracoscopic-assisted Nuss operation was completed by bilateral incisions. The operation time, intraoperative blood loss, and postoperative hospitalization were counted and analyzed. The satisfaction of the surgery was evaluated according to the Haller index and the most posterior sternal compression sternovertebral distance. RESULTS: The surgeries were successfully completed in 10 patients without a transfer to open procedure. The average operation time was (56 ± 8.76) min, the intraoperative blood loss was (23.5 ± 11.07) mL, and the postoperative hospitalization was (7.2 ± 0.92) d. There were no serious complications or death during the perioperative period. Compared with the data before the operation, the most posterior sternal compression sternovertebral distance was larger, and the Haller index was lower, the differences were statistically significant (P < 0.05). CONCLUSIONS: 3D reconstruction technology combined with 3D printing, which can be used before operation, contributes to the operator performing thoracoscopic-assisted Nuss operation safely and effectively, which has productive clinical application value for the treatment of pectus excavatum.

13.
Eur J Pharmacol ; 928: 175114, 2022 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-35764130

RESUMO

High temperature requirement A3 (HtrA3) belongs to the HtrA family, and its role in inflammation and myocardial ischemia-reperfusion injury remains unknown. Herein, the study aimed to explore the role of HtrA3 in inflammatory cytokine secretion and the nuclear factor kappa B (NF-κB) signaling pathway in hypoxia-reoxygenation (H/R)-induced H9C2 cardiomyoblasts. H9C2 cells were treated with H/R to mimic myocardial ischemia-reperfusion in vitro. Results showed that HtrA3 expression was significantly downregulated and the expression of inflammatory cytokines was regulated in response to H/R. HtrA3 overexpression decreased the secretion of inflammatory cytokines, whereas HtrA3 knockdown led to increase levels of inflammatory cytokines. And H/R-induced inflammation in H9C2 cells was inhibited by the regulation of the NF-κB signaling pathway. Our findings demonstrate that HtrA3 alleviates H/R-induced inflammatory responses in H9C2 cardiomyoblasts, possibly by suppressing the pro-inflammatory NF-κB signaling pathway.


Assuntos
Hipóxia , NF-kappa B , Serina Endopeptidases , Animais , Apoptose , Citocinas/metabolismo , Hipóxia/metabolismo , Inflamação/metabolismo , Miócitos Cardíacos/metabolismo , NF-kappa B/metabolismo , Ratos , Serina Endopeptidases/metabolismo
14.
Am J Transl Res ; 14(5): 2910-2925, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35702071

RESUMO

OBJECTIVE: To evaluate the biological properties of modified 3D printing scaffold (PTS) and applied the hybrid graft for in situ transplantation. METHODS: PTS was prepared via 3D printing and modified by Pluronic F-127. Biocompatibility of the scaffold was examined in vitro to ascertain its benefit in attachment and proliferation of bone marrow mesenchymal stem cells (BMSCs). Moreover, a hybrid trachea was constructed by combining the modified PTS with decellularized matrix. Finally, two animal models of in situ transplantation were established, one for repairing tracheal local window-shape defects and the other for tracheal segmental replacement. RESULTS: The rough surface and chemical elements of the scaffold were improved after modification by Pluronic F-127. Results of BMSCs inoculation showed that the modified scaffold was beneficial to attachment and proliferation. The epithelial cells were seen crawling on and attaching to the patch, 30 days following prothetic surgery of the local tracheal defects. Furthermore, the advantages of the modified PTS and decellularized matrix were combined to generate a hybrid graft, which was subsequently applied to a tracheal segmental replacement model. CONCLUSION: Pluronic F-127-based modification generated a PTS with excellent biocompatibility. The modified scaffold has great potential in development of future therapies for tracheal replacement and reconstruction.

15.
Curr Drug Deliv ; 19(9): 928-939, 2022 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-35473528

RESUMO

BACKGROUND: Iron is used to alter macrophage phenotypes and induce tumor cell death. Iron oxide nanoparticles can induce macrophage polarization into the M1 phenotype, which inhibits tumor growth and can dissociate into iron ions in macrophages. OBJECTIVE: In this study, we proposed to construct high expression of Ferroportin1 macrophages as carriers to deliver Fe3O4-nanoparticles and iron directly to tumor sites. METHODS: Three sizes of Fe3O4-nanoparticles with gradient concentrations were used. The migration ability of iron-carrying macrophages was confirmed by an in vitro migration experiment and monocyte chemoattractant protein-1 detection. The release of iron from macrophages was confirmed by determining their levels in the cell culture supernatant, and we constructed a high expression of ferroportin strain of macrophage lines to increase intracellular iron efflux by increasing membrane transferrin expression. Fe3O4-NPs in Ana-1 cells were degraded in lysosomes, and the amount of iron released was correlated with the expression of ferroportin1. RESULTS: After Fe3O4-nanoparticles uptake by macrophages, not only polarized macrophages into M1 phenotype, but the nanoparticles also dissolved in the lysosome and iron were released out of the cell. FPN1 is the only known Fe transporter; we use a Lentiviral vector carrying the FPN1 gene transfected into macrophages, has successfully constructed Ana-1-FPN1 cells, and maintains high expression of FPN1. Ana-1-FPN1 cells increase intracellular iron release. Fe3O4-nanoparticles loaded with engineered Ana-1 macrophages can act as a "reservoir" of iron. CONCLUSION: Our study provides proof of strategy for Fe3O4-NPs target delivery to the tumor microenvironment. Moreover, increase of intracellular iron efflux by overexpression of FPN1, cell carriers can act as a reservoir for iron, providing the basis for targeted delivery of Fe3O4-NPs and iron ions in vivo.


Assuntos
Ferro , Nanopartículas , Ferro/metabolismo , Macrófagos , Microambiente Tumoral
17.
J Mater Chem B ; 10(25): 4810-4822, 2022 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-35237780

RESUMO

Long segment trachea defects are repaired by tracheal substitution, while decellularized technology has been effectively employed to prepare tissue engineering trachea (TET). However, its clinical application is restricted by the long preparation cycle, while poor vascularization is associated with the transplantation failure. In the present study, we used sodium lauryl ether sulfate (SLES) to develop a novel rapid decellularized tracheal preparation method, then constructed a TET with revascularization functions. Summarily, we decellularized rabbit trachea using various SLES concentrations. Results from histological analysis, immunohistochemical and DAPI staining, as well as DNA quantitative assay, revealed that 1-0.1% (v/v) SLES treatment not only entirely removed cellular components to reduce its immunogenicity, but also retained the tracheal matrix's gross structure. SEM images, safranine O-fast green staining, total collagen content assay and collagen II immunofluorescence revealed that low SLES concentrations preserved the bioactive components of the decellularized tracheal matrix. Next, we performed cytobiocompatible and cytotoxin assays to verify biocompatibility of the decellularized tracheal matrix, and is confirmed by the omentum transplantation of rats. Results from omentum transplantation revealed that the decellularized tracheal matrix had low immunogenicity and excellent biocompatibility. Its revascularization capacity was confirmed by histologic appearance and CD31 immunofluorescence. Based on these findings, we selected 0.1% (v/v) as the optimal SLES concentration for preparing a decellularized tracheal matrix. Next, we seeded allogeneic bone marrow stem cells (BMSC) onto the matrix to construct TET patches. In vivo tracheal defect reconstruction confirmed the biocompatibility and revascularization capacity of this novel TET, and the formation of a vascular network around the patch promoted submucosa and mucosa regeneration without significant stenosis, 4 weeks post-surgery. In conclusion, we used SLES to successfully develop a novel decellularized approach for the preparation of TET, which has low immunogenic and inflammatory responses, as well as excellent biocompatibility, and revascularization ability in vivo without additional exogenous cytokines.


Assuntos
Engenharia Tecidual , Traqueia , Animais , Matriz Extracelular/química , Coelhos , Ratos , Dodecilsulfato de Sódio , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Traqueia/fisiologia , Traqueia/cirurgia
18.
J Biomed Mater Res A ; 110(5): 1062-1076, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35064753

RESUMO

The construction of ideal tissue engineering trachea has always been a difficult problem in trachea transplantation surgery. The biological characteristics of decellularized matrix prepared by detergent-enzymatic (DEM) and 3D printing biomimetic scaffold (PTS) in vivo and in vitro were compared. In order to comprehensively evaluate its performance, we tested morphological and biomechanical characteristics of the native tracheas(Group A), DEM(Group B), and PTS(Group C). The above groups were co-cultured with bone marrow mesenchymal stem cells (BMSCs), after which cell attachment and proliferation on the scaffolds were detected. Allogeneic implantation experiments were performed to assess the in vivo biocompatibility of the studied scaffolds. Moreover, an in-situ experiment of the tracheal repair was conducted to compare the survival of every group. The biomechanical properties of PTS were significantly better than those of other scaffolds (P < .05). And they retained their structural integrity in the host compared with the other scaffolds (P < .05). Besides, significantly milder immune-rejection reactions were observed in Group C than those in Group A (P < .05). In situ experiments showed that Group C significantly a good postoperative condition compared with the other scaffold groups (P < .05). Fiberoptic bronchoscopy analysis of PTS showed a better condition in the lumen. In conclusion, PTS has excellent biomechanical properties. Although the PTS group showed lower biocompatibility than the decellularized group, it exhibited better cell attachment and proliferation. In situ transplantation results showed that PTS could be an ideal source of tissue engineering material for tracheal repair.


Assuntos
Alicerces Teciduais , Traqueia , Biomimética , Matriz Extracelular/química , Impressão Tridimensional , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Traqueia/química
19.
Cell Cycle ; 20(21): 2233-2247, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34499581

RESUMO

Magnesium transporter subtype 1 (MAGT1) is known to participate in animal development and cell differentiation. Thus far, MAGT1 studies have mainly focused on its role in cardiomyocyte regulation and differentiation; only a few studies have demonstrated its role in cell proliferation. To investigate the underlying mechanism of MAGT1 in cell proliferation, HeLa and SiHa cells were transiently knocked down with different siRNAs. We showed that cell proliferation was substantially restricted by S-phase arrest and apoptosis in the MAGT1-knocked down cells, which was further confirmed by the increased expression of p21, cyclin-A1, and cyclin-B1, as well as the decreased expression of MYC, cyclin-D1, cyclin-E1, and CDK2. MAGT1 knockdown also resulted in significant changes in the transcriptional expression of 1,598 genes that were analyzed by RNA sequencing. Bioinformatics analysis showed that MAGT1 was related to the MAPK signaling pathway. Western blot analysis confirmed that the phosphorylation of extracellular signal-related protein kinase 1/2 (ERK1/2) and p38 was remarkably reduced in MAGT1 down-regulated groups. Additionally, MAGT1 was required for the function of viral proteins E6/E7 during cell proliferation and G1/S cell-cycle progression. Therefore, MAGT1 plays a crucial role in the proliferation of HPV-positive cervical cancer cells, S-phase progression, and the ERK/p38 MAPK signaling pathway. These results indicate the potential of MAGT1 as a novel target for anticancer research.Abbreviations: MAGT1: Magnesium transporter subtype 1; MAPK: Mitogen-activated protein kinase; XMEN: X-linked immunodeficiency with Magnesium defect, Epstein-Barr virus infection and Neoplasia; BMMSCs: Bone Marrow Mesenchymal Stem Cells; Dpp: Decapentaplegic; CDKIs: CDK inhibitors; GPCR: G-protein coupled receptor; GO: Gene Ontology; KEGG: Kyoto Encyclopedia of Genes and Genomes; RTK: Receptor Tyrosine Kinase; PTK: Protein Tyrosine Kinase; FGFR: Fibroblast Growth Factor Receptor; BMP: Bone Morphogenetic Protein; HPV18 E6/E7: Human Papillomavirus 18 Early protein 6/ early protein 7; FACS: Fluorescence Activated Cell Sorting; PI: Propidium Iodide.


Assuntos
Proteínas de Transporte de Cátions , Infecções por Vírus Epstein-Barr , Animais , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Células HeLa , Herpesvirus Humano 4/metabolismo , Humanos , Proteínas Proto-Oncogênicas c-myc , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
20.
Mater Sci Eng C Mater Biol Appl ; 128: 112201, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34474813

RESUMO

Tissue engineering technology provides effective alternative treatments for tracheal reconstruction. The formation of a functional microvascular network is essential to support cell metabolism and ensure the long-term survival of grafts. However, given the lack of an identifiable vascular pedicle of the trachea that could be anastomosed to the blood vessels directly in the recipient's neck, successful tracheal transplantation faces significant challenges in rebuilding the adequate blood supply of the graft. Herein, we describe a one-step method to construct microvascularization of tissue-engineered trachea in orthotopic transplantation. Forty rabbit tracheae were decellularized using a vacuum-assisted decellularization (VAD) method. Histological appearance and immunohistochemical (IHC) analysis demonstrated efficient removal of cellular components and nuclear material from natural tissue, which was also confirmed by 4'-6-diamidino-2-phenylindole(DAPI) staining and DNA quantitative analysis, thus significantly reducing the antigenicity. Scanning electron microscopy (SEM), immunofluorescence (IF) analysis, GAG and collagen quantitative analysis showed that the hierarchical structures, composition and integrity of the extracellular matrix (ECM) were protected. IF analysis also demonstrated that basic fibroblast growth factor (b-FGF) was preserved during the decellularization process, and also exerted biocompatibility and proangiogenic properties by the chick chorioallantoic membrane(CAM) assay. Xenotransplantation assays indicated that the VAD tracheal matrix would no longer induced inflammatory reactions implanted in the body for 4 weeks after treated by VAD more than 16 h. Furthermore, we seeded the matrix with bone marrow-derived endothelial cells (BMECs) in vitro and performed in vivo tracheal patch repair assays to prove the biocompatibility and neovascularization of VAD-treated tracheal matrix, and the formation of a vascular network around the patch promoted the crawling of surrounding ciliated epithelial cells to the surface of the graft. We conclude that this natural VAD tracheal matrix is non-immunogenic and no inflammatory reactions in vivo transplantation. Seeding with BMECs on the grafts and then performing orthotopic transplantation can effectively promote the microvascularization and accelerate the native epithelium cells crawling to the lumen of the tracheal graft.


Assuntos
Engenharia Tecidual , Traqueia , Animais , Células Endoteliais , Matriz Extracelular , Coelhos , Alicerces Teciduais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...