Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Heliyon ; 10(1): e23705, 2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-38192833

RESUMO

Background: It remains undetermined whether preoperative computed tomography (CT)-guided hookwire localization would result in elevated risk of tumor spread through air spaces (STAS) in stage IA lung adenocarcinoma. Methods: A total of 1836 patients who underwent lobectomy were included. To eliminate the potential impact of confounding factors on producing STAS, propensity score-matching (PSM) was used to create two balanced subgroups stratified by implementation of hookwire localization. We also introduced an external cohort including 1486 patients to explore the effect of hookwire localization on the incidence of STAS and patient survival after sublobar resection (SR). For proactive simulation of hookwire localization, 20 consecutive lobectomy specimens of p-stage IA lung adenocarcinoma were selected. Results: Ex vivo tests revealed that mechanical artifacts presenting as spreading through a localizer surface (STALS) could be induced by hookwire localization but be distinguished by CD68 and AE1/3 antibody-based immunohistochemistry. The distance of STALS dissemination tended to be shorter compared with real STAS (P = 0.000). After PSM, implementation of hookwire localization was not associated with elevated STAS incidence, nor worse survival in p-stage IA patients undergoing lobectomy irrespective of STAS. Conclusions: CT-guided hookwire localization might induce mechanical artifacts presenting as STALS which could be distinguished by immunohistochemistry, but would not affect survival in p-stage IA disease. Surgeons can be less apprehensive about performing hookwire localization in relation to STAS on stage IA disease suitable for SR.

2.
Thorac Cancer ; 15(4): 286-298, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38151913

RESUMO

BACKGROUND: GTPase immunity-associated protein 7 (GIMAP7) has been previously recognized as a prognostic marker in pan-cancer. Our objective was to explore the function of GIMAP7 in the progression of lung adenocarcinoma (LUAD). METHODS: GIMAP7 was overexpressed by transfection with GIMAP7 plasmid, and knocked down using siRNAs. The biological functions of GIMAP7 were examined by employing CCK-8, EdU, colony formation, flow cytometry, wound healing, and transwell assays. The effects of GIMAP7 on the extracellular acidification rate (ECAR), oxygen consumption rate (OCR), lactate production, and glucose uptake were evaluated. In addition, the related mRNA and protein expression was detected employing immunohistochemical, western blot, and qRT-PCR. A xenograft tumor model was established in nude mice to evaluate the effects of GIMAP7 on tumor growth. RESULTS: GIMAP7 was lowly expressed in LUAD tissues and cells. GIMAP7 inhibited the proliferation, mobility, EMT, glycolysis, but promoted apoptosis in LUAD cells. Moreover, we also confirmed that GIMAP7 suppressed Smo/AMPK signaling in LUAD cells. By adding the Smo agonist SAG and AMPK agonist GSK621, the results of rescue experiments further verified that GIMAP7 played a role in LUAD inhibition through inhibition of the Smo/AMPK signaling pathway. Furthermore, the role of GIMAP7 in inhibiting tumor growth was verified in vivo. CONCLUSIONS: These results demonstrate that GIMAP7 could inhibit cell proliferation, mobility and glycolysis, but accelerate apoptosis via repressing the Smo/AMPK signaling pathway in LUAD.


Assuntos
Adenocarcinoma de Pulmão , Neoplasias Pulmonares , Animais , Camundongos , Humanos , GTP Fosfo-Hidrolases , Proteínas Quinases Ativadas por AMP , Camundongos Nus , Transdução de Sinais , Transição Epitelial-Mesenquimal , Adenocarcinoma de Pulmão/genética , Glicólise , Proliferação de Células , Modelos Animais de Doenças , Neoplasias Pulmonares/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Proteínas de Ligação ao GTP
3.
Aging (Albany NY) ; 15(21): 11970-11984, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37910672

RESUMO

BACKGROUND: Lung adenocarcinoma accounts for approximately 40% of lung cancer cases and poses a serious threat to human health. Therefore, there is an urgent need to identify central biomarkers in lung adenocarcinoma. METHODS: We first identified the EMT-associated genes in LUAD based on the TCGA cohort. Then we screened these 90 EMT-associated genes using univariate Cox regression analysis and LASSO regression analysis to develop a prognostic gene signature in the training set. The predictive performance of the gene signature was assessed in the validation set and multiple external test sets using the ROC cure, C index and log-rank tests. RT-PCR, western blot, wound healing assays, and siRNA methods were further used to investigate the role of PLEK2 in tumor behaviors. RESULTS: Eight genes (CCNB1, PLEK2, DERL3, C1QTNF6, DLGAP5, HMMR, GJB3, and SPOCK1) were eventually selected to develop an eight-gene signature. The 5-year AUC of the gene signature has a robust predictive ability both for predicting overall survival (0.774, 0.756, and 0.669 in the external test sets, respectively), and for progression free survival (0.774, 0.746, and 0.755 in the external test sets, respectively). C-index of the gene signature was 0.961 ± 0.005, 0.916 ± 0.011, and 0.868 ± 0.234 in the external test sets, respectively. Four genes (C1QTNF6, DLGAP5, HMMR, and PLEK2) were identified as key genes in LUAD progression, which were upregulated in the cancerous tissue compared with in the normal tissue (P < 0.001), and correlated with an unwanted prognosis in lung cancer (P < 0.05). PLEK2 was used as an example to explore its effect on LUAD progression in vitro using RT-PCR, western blot, CCK8, si-RNA and wound healing assay. Silencing of PLEK2 was shown to reduce proliferative and migrated ability of lung cancer cells via prohibition of autophagy. CONCLUSIONS: This study developed a novel EMT-related gene signature benefiting precision medicine, and identified four pivotal genes which can serve as therapeutic targets in LUAD. Four key genes can serve as molecular targets for patients with LUAD; silencing of PLEK2 was shown to reduce proliferative and migrated ability of lung cancer cells via prohibition of autophagy.


Assuntos
Adenocarcinoma de Pulmão , Neoplasias Pulmonares , Humanos , Transição Epitelial-Mesenquimal/genética , Adenocarcinoma de Pulmão/genética , Neoplasias Pulmonares/genética , Biomarcadores , Biologia Computacional , Prognóstico , Proteoglicanas , Proteínas de Membrana
4.
Small Methods ; 7(7): e2300096, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37086121

RESUMO

The phenotypic changes of circulating tumor cells (CTCs) during the epithelial-mesenchymal transition (EMT) have been a hot topic in tumor biology and cancer therapeutic development. Here, an integrated platform of single-cell fluorescent enzymatic assays with superwetting droplet-array microchips (SDAM) for ultrasensitive functional screening of epithelial-mesenchymal sub-phenotypes of CTCs is reported. The SDAM can generate high-density, volume well-defined droplet (0.66 nL per droplet) arrays isolating single tumor cells via a discontinuous dewetting effect. It enables sensitive detection of MMP9 enzyme activities secreted by single tumor cells, correlating to their epithelial-mesenchymal sub-phenotypes. In the pilot clinical double-blind tests, the authors have demonstrated that SDAM assays allow for rapid identification and functional screening of CTCs with different epithelial-mesenchymal properties. The consistency with the clinical outcomes validates the usefulness of single-cell secreted MMP9 as a biomarker for selective CTC screening and tumor metastasis monitoring. Convenient addressing and recovery of individual CTCs from SDAM have been demonstrated for gene mutation sequencing, immunostaining, and transcriptome analysis, revealing new understandings of the signaling pathways between MMP9 secretion and the EMT regulation of CTCs. The SDAM approach combined with sequencing technologies promises to explore the dynamic EMT plasticity of tumors at the single-cell level.


Assuntos
Transição Epitelial-Mesenquimal , Células Neoplásicas Circulantes , Humanos , Biomarcadores Tumorais/metabolismo , Transição Epitelial-Mesenquimal/genética , Metaloproteinase 9 da Matriz/genética , Células Neoplásicas Circulantes/metabolismo , Método Duplo-Cego
5.
Cancer Med ; 12(8): 9547-9558, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36880167

RESUMO

AIMS: We investigated the clinical implications and molecular features of TLS in stage I lung adenocarcinoma (LUAD). METHODS: We retrospectively reviewed the clinicopathological characteristics of 540 patients with p-stage I LUAD. Logistic regression analysis was applied to determining the relationships between clinicopathological features and the presence of TLS. TLS-associated immune infiltration pattern and signature genes were characterized using the transcriptomic profiles of 511 LUADs from The Cancer Genome Atlas (TCGA) database. RESULTS: The presence of TLS was associated with a higher pT stage, low- and middle-grade patterns, and the absence of tumor spreading through air spaces (STAS) and subsolid nodules. Multivariate Cox regression analysis identified that the presence of TLS was associated with favorable overall survival (OS) (p < 0.001) and recurrence-free survival (RFS) (p < 0.001). Subgroup analysis showed that the most favorable OS (p < 0.001) and RFS (p < 0.001) favored the TLS + PD-1- subgroup. The presence of TLS was characterized by abundance in antitumor immunocytes including activated CD8+ T and B cells as well as dentritic cells in TCGA cohort. CONCLUSION: The presence of TLS was an independent favorable factor for patients with stage I LUAD. The presence TLS was featured by special immune profiles which might aid oncologists in determining personalized adjuvant treatment.


Assuntos
Adenocarcinoma de Pulmão , Neoplasias Pulmonares , Estruturas Linfoides Terciárias , Humanos , Estudos Retrospectivos , Estruturas Linfoides Terciárias/patologia , Prognóstico , Adenocarcinoma de Pulmão/patologia , Neoplasias Pulmonares/patologia
6.
Adv Mater ; 35(23): e2210691, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36913720

RESUMO

siRNA-mediated management of myocardial ischemia reperfusion (IR) injury is greatly hampered by the inefficient myocardial enrichment and cardiomyocyte transfection. Herein, nanocomplexes (NCs) reversibly camouflaged with a platelet-macrophage hybrid membrane (HM) are developed to efficiently deliver Sav1 siRNA (siSav1) into cardiomyocytes, suppressing the Hippo pathway and inducing cardiomyocyte regeneration. The biomimetic BSPC@HM NCs consist of a cationic nanocore assembled from a membrane-penetrating helical polypeptide (P-Ben) and siSav1, a charge-reversal intermediate layer of poly(l-lysine)-cis-aconitic acid (PC), and an outer shell of HM. Due to HM-mediated inflammation homing and microthrombus targeting, intravenously injected BSPC@HM NCs can efficiently accumulate in the IR-injured myocardium, where the acidic inflammatory microenvironment triggers charge reversal of PC to shed off both HM and PC layers and allow the penetration of the exposed P-Ben/siSav1 NCs into cardiomyocytes. In rats and pigs, BSPC@HM NCs remarkably downregulates Sav1 in IR-injured myocardium, promotes myocardium regeneration, suppresses myocardial apoptosis, and recovers cardiac functions. This study reports a bioinspired strategy to overcome the multiple systemic barriers against myocardial siRNA delivery, and holds profound potential for gene therapy against cardiac injuries.


Assuntos
Traumatismo por Reperfusão Miocárdica , Ratos , Suínos , Animais , Traumatismo por Reperfusão Miocárdica/terapia , Traumatismo por Reperfusão Miocárdica/metabolismo , RNA Interferente Pequeno/metabolismo , Biomimética , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Apoptose
7.
Eur J Cardiothorac Surg ; 63(3)2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36825852

RESUMO

OBJECTIVES: This study aimed to investigate the potential of complex glandular patterns (CGP) in lymph node micrometastasis (LNMM) and to determine the clinical beneficiaries in stage I lung adenocarcinoma (LUAD) with CGP. Meanwhile, the feasibility of detecting CGP on frozen section was also evaluated. METHODS: We retrospectively analysed the clinicopathological characteristics of 848 pathologic-stage I LUADs. A logistic regression model and a Cox proportional-hazards model were conducted to define the risk factors for LNMM and survival respectively. Furthermore, 5 pathologists reviewed frozen sections of 100 LUADs independently. RESULTS: The logistic regression model indicated that CGP [odds ratio 3.89, 95% confidence interval (CI) 2.46-6.15; P < 0.001] was an independent predictor of the presence of LNMM. Subgroup analysis revealed that CGP-present/LNMM-positive LUAD had the highest risk of both loco-regional and distant recurrence. Moreover, adequate lymphadenectomy [recurrence-free survival: hazard ratio (HR) 0.61, 95% CI 0.40-0.95; P = 0.028; overall survival: HR 0.64, 95% CI 0.41-0.99; P = 0.043] and adjuvant chemotherapy (recurrence-free survival: HR 0.30, 95% CI 0.18-0.52; P < 0.001; overall survival: HR 0.33, 95% CI 0.19-0.57; P < 0.001) brought survival benefits to CGP-present patients, especially to CGP-present/LNMM-positive subgroup. Across the 5 pathologists, sensitivity ranged from 59 to 68% and specificity ranged from 79 to 83%, with moderate diagnostic agreement and high interobserver agreement for detecting CGP on frozen section. CONCLUSIONS: LNMM was more frequently observed in stage I LUAD with CGP. Adequate lymphadenectomy and adjuvant chemotherapy were associated with improved survival in CGP-present patients, especially in CGP-present/LNMM-positive subgroup. Additionally, it is feasible to identify CGP on frozen section intraoperatively.


Assuntos
Adenocarcinoma de Pulmão , Neoplasias Pulmonares , Humanos , Prognóstico , Estudos Retrospectivos , Metástase Linfática , Adenocarcinoma de Pulmão/cirurgia , Adenocarcinoma de Pulmão/patologia , Micrometástase de Neoplasia/patologia , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/cirurgia , Neoplasias Pulmonares/patologia , Estadiamento de Neoplasias
8.
Biomater Sci ; 10(21): 6258-6266, 2022 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-36111777

RESUMO

Apoptosis of cardiomyocytes is a critical outcome of myocardial ischemia-reperfusion injury (MIRI), which leads to the permanent impairment of cardiac function. Upregulated E2F1 is implicated in inducing cardiomyocyte apoptosis, and thus intervention of the E2F1 signaling pathway via RNA interference may hold promising potential for rescuing the myocardium from MIRI. To aid efficient E2F1 siRNA (siE2F1) delivery into cardiomyocytes that are normally hard to transfect, a spherical, α-helical polypeptide (SPP) with potent membrane activity was developed via dendrimer-initiated ring-opening polymerization of N-carboxyanhydride followed by side-chain functionalization with guanidines. Due to its multivalent structure, SPP outperformed its linear counterpart (LPP) to feature potent siRNA binding affinity and membrane activity. Thus, SPP effectively delivered siE2F1 into cardiomyocytes and suppressed E2F1 expression both in vitro and in vivo after intramyocardial injection. The E2F1-miR421-Pink1 signaling pathway was disrupted, thereby leading to the reduction of MIRI-induced mitochondrial damage, apoptosis, and inflammation of cardiomyocytes and ultimately recovering the systolic function of the myocardium. This study provides an example of membrane-penetrating nucleic acid delivery materials, and it also provides a promising approach for the genetic manipulation of cardiomyocyte apoptosis for the treatment of MIRI.


Assuntos
Dendrímeros , Traumatismo por Reperfusão Miocárdica , Ratos , Animais , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , RNA Interferente Pequeno/farmacologia , Dendrímeros/metabolismo , Ratos Sprague-Dawley , Apoptose , Peptídeos/genética , Peptídeos/farmacologia , Peptídeos/metabolismo , Guanidinas/farmacologia , Guanidinas/uso terapêutico , Proteínas Quinases/metabolismo , Proteínas Quinases/farmacologia , Proteínas Quinases/uso terapêutico , Fator de Transcrição E2F1/metabolismo , Fator de Transcrição E2F1/farmacologia
9.
Zhongguo Fei Ai Za Zhi ; 25(9): 642-650, 2022 Sep 20.
Artigo em Chinês | MEDLINE | ID: mdl-36172728

RESUMO

BACKGROUND: Patients who underwent lobectomy resection are prone to hypoxemia, and the vast majority present with type I respiratory failure. Thus, improvement of hypoxemia is one of the most important factors to facilitate postoperative recovery of patients. In this study, the superiority-inferiority of different oxygen inhalation methods were compared with high-flow nasal oxygen therapy (HFNO), noninvasive mechanical ventilation (NIMV) and nasal oxygen breath (NOB) in patients with hypoxemia after single-port video-assisted thoracoscopic (VATS) lobectomy, and the clinical efficacy of HFNO in these patients was further investigated. METHODS: A total of 180 patients from the Second Affiliated Hospital of Soochow University in China with hypoxemia who accepting single-port VATS lobectomy from June 2021 to March 2022 were randomly divided into three groups (n=60), which were treated with HFNO, NIMV and NOB, respectively. The results of arterial blood gas analysis, patient's comfort score and incidence of complications were observed before, 1 h, 6 h-12 h and after use. Statistical analyses were conducted using statistical program for social sciences 25.0 (SPSS 25.0), and P<0.05 was considered as statistical significance. RESULTS: For patients with hypoxemia after accepting single-port VATS lobectomy, HFNO was no less effective than NIMV (P=0.333), and both of whom could fast increase patients' partial pressure of oxygen/fraction of inspiration O2 (PaO2/FiO2) compared to NOB (P<0.001). Besides, HFNO shows a great advantage in comfort degree and stay length (P<0.001, P=0.004), and incidence of complications were slightly lower than other groups (P=0.232). But it is worthy to note that HFNO is still slightly less effective than NIMV in patients with postoperative hypoxemia accompanied by elevated partial pressure of carbon dioxide (PaCO2). CONCLUSIONS: For patients with hypoxemia who accepting single-port VATS lobectomy, HFNO can be used as the first choice. However, for patients with postoperative hypoxemia accompanied by elevated PaCO2, NIMV is still recommended to improve oxygenation.


Assuntos
Neoplasias Pulmonares , Cirurgia Torácica Vídeoassistida , Dióxido de Carbono , Humanos , Hipóxia/etiologia , Hipóxia/cirurgia , Neoplasias Pulmonares/cirurgia , Oxigênio , Pneumonectomia/métodos , Cirurgia Torácica Vídeoassistida/métodos
10.
Thorac Cancer ; 13(19): 2767-2775, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36054738

RESUMO

BACKGROUND: The significance of postoperative adjuvant radiotherapy (PORT) on the survival of resected IIIA-N2 non-small cell lung cancer (NSCLC) remains controversial. Here, we aimed to determine the predictive value of the three nodal classifications which might aid in PORT decision-making. METHODS: A total of 4797 patients with stage IIIA-N2 resected NSCLC were identified in the Surveillance, Epidemiology and End Results (SEER) database and were grouped by whether PORT was administered. Survival analysis was used to identify the patient groups who can benefit from PORT. Multivariate analysis was performed to confirm the independent risk factors for lung cancer-specific survival (LCSS) and overall survival (OS). A validation cohort of 1184 patients from three medical centers in China were also included. RESULTS: PORT was not associated with better LCSS and OS in the entire cohort after propensity score matching (PSM). However, in the subgroups of positive lymph nodes 4 (PLN4), lymph node ratio 4 (LNR4), and log odds of positive lymph nodes 4 (LODDS4), PORT exhibited its role in improving LCSS (p < 0.05). Although the three nodal classifications were all identified as independent predictors of LCSS and OS, LODDS classification had the best discriminatory ability and prognostic accuracy for stage IIIA-N2 patients. Similar results were also obtained in the validation cohort. CONCLUSIONS: The LODDS classification not only exhibited the best prognostic performance in predicting LCSS and OS in stage IIIA-N2 disease, but also could help tailor individualized PORT.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Neoplasias Testiculares , Carcinoma Pulmonar de Células não Pequenas/radioterapia , Carcinoma Pulmonar de Células não Pequenas/cirurgia , Humanos , Neoplasias Pulmonares/radioterapia , Neoplasias Pulmonares/cirurgia , Linfonodos/patologia , Linfonodos/cirurgia , Masculino , Estadiamento de Neoplasias , Prognóstico , Radioterapia Adjuvante , Neoplasias Testiculares/patologia
12.
Bioact Mater ; 17: 320-333, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35386446

RESUMO

Ischemia-reperfusion (IR) injury represents a major cause of myocardial dysfunction after infarction and thrombolytic therapy, and it is closely related to the free radical explosion and overwhelming inflammatory responses. Herein, macrophage-targeting nanocomplexes (NCs) are developed to mediate efficient co-delivery of siRNA against MOF (siMOF) and microRNA-21 (miR21) into myocardial macrophages, cooperatively orchestrating the myocardial microenvironment against IR injury. Bioreducible, branched poly(ß-amino ester) (BPAE-SS) is designed to co-condense siMOF and miR21 into NCs in a multivalency-reinforced approach, and they are surface-decorated with carboxylated mannan (Man-COOH) to shield the positive surface charges and enhance the serum stability. The final MBSsm NCs are efficiently internalized by myocardial macrophages after systemic administration, wherein BPAE-SS is degraded into small segments by intracellular glutathione to promote the siMOF/miR21 release, finally provoking efficient gene silencing. Thus, cardiomyocyte protection and macrophage modulation are realized via the combined effects of ROS scavenging, inflammation inhibition, and autophagy attenuation, which ameliorates the myocardial microenvironment and restores the cardiac function via positive cellular crosstalk. This study renders promising solutions to address the multiple systemic barriers against in vivo nucleic acid delivery, and it also offers new options for IR injury by manipulating multiple reciprocal bio-reactions.

13.
Adv Mater ; 34(23): e2109517, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35388551

RESUMO

Systemic immunosuppression mediated by tumor-derived exosomes is an important cause for the resistance of immune checkpoint blockade (ICB) therapy. Herein, self-adaptive platelet (PLT) pharmacytes are engineered to mediate cascaded delivery of exosome-inhibiting siRNA and anti-PD-L1 (aPDL1) toward synergized antitumor immunity. In the pharmacytes, polycationic nanocomplexes (NCs) assembled from Rab27 siRNA (siRab) and a membrane-penetrating polypeptide are encapsulated inside the open canalicular system of PLTs, and cytotoxic T lymphocytes (CTLs)-responsive aPDL1 nanogels (NGs) are covalently backpacked on the PLT surface. Upon systemic administration, the pharmacytes enable prolonged blood circulation and active accumulation to tumors, wherein PLTs are activated to liberate siRab NCs, which efficiently transfect tumor cells, silence Rab27a, and inhibit exosome secretion. The immunosuppression is thus relieved, leading to the activation, proliferation, and tumoral infiltration of cytotoxic T cells, which trigger latent aPDL1 release. As such, the competitive aPDL1 exhaustion by PD-L1-expressing exosomes is minimized to sensitize ICB. Synergistically, siRab and aPDL1 induce strong antitumor immunological response and memory against syngeneic murine melanoma. This study reports a bioinspired mechanism to resolve the blood circulation/cell internalization contradiction of polycationic siRNA delivery systems, and renders an enlightened approach for the spatiotemporal enhancement of antitumor immunity.


Assuntos
Imunoterapia , Melanoma , Animais , Linhagem Celular Tumoral , Melanoma/patologia , Camundongos , RNA Interferente Pequeno/genética , Linfócitos T Citotóxicos , Microambiente Tumoral
14.
Ann Transl Med ; 10(1): 12, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35242857

RESUMO

BACKGROUND: Anatomical segmentectomy by uniportal video-assisted thoracoscopic surgery (U-VATS) is a delicate surgical procedure. Hitherto, only few studies have assessed the learning curves of anatomical segmentectomy by U-VATS, with varying data available. The present study aimed to investigate the learning curve and clinical advantages for U-VATS segmentectomy. METHODS: The medical records of patients who underwent U-VATS or non-U-VATS segmentectomy between August 2017 and May 2020 were retrospectively reviewed. Cumulative sum (CUSUM) analysis was employed to illustrate the learning curve of U-VATS segmentectomy. Perioperative parameters were used to determine the structural intervals of the learning curve, and to compare U-VATS and non-U-VATS segmentectomy. RESULTS: In total, 122 patients receiving U-VATS segmentectomy and 98 patients receiving non-VATS segmentectomy were included. Of these, 116 patients underwent successful U-VATS segmentectomy, while the other six patients underwent conversions. The structural intervals of 20-29 cases and 58-63 cases were determined as the threshold according to the CUSUM analyses. The learning process of U-VATS segmentectomy was therefore divided into three phases. Interestingly, the perioperative parameters differed significantly between Phases 1 and 3, including operative time (Op-T), postoperative hospital stays (Po-Hst), postoperative thoracic drainage (Po-D), and operative failure (Po-F) rates (P<0.05). Moreover, U-VATS segmentectomy in Phase 3 was associated with significantly shorter Po-Hst and Op-T, less Po-D, and reduced postoperative pain compared with non-U-VATS (P<0.05). CONCLUSIONS: U-VATS segmentectomy is an ideal alternative to non-U-VATS segmentectomy. Surgeons can preliminarily complete U-VATS anatomical segmentectomy after performing 20-29 cases, and can master the surgical techniques after completing 58-63 cases.

15.
Aging (Albany NY) ; 13(19): 23376-23392, 2021 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-34644678

RESUMO

Lung adenocarcinoma (LUAD) is the most common histological type of non-small cell lung cancer (NSCLC). Due to the nonspecific early symptoms, the majority of the diagnosed LUAD patients are in the middle and late stages, with multiple metastases, and have missed the optimal period for treatment. Current studies have reported lncRNA DARS-AS1 as a cancer-promoting gene that expedites tumorigenesis. This is the first study demonstrating that DARS-AS1 is involved in the mediating process of LUAD. Cell functional experiments revealed that lncRNA DARS-AS1 participated in enhancing LUAD proliferation, invasion, and migration by inhibiting miR-188-5p. The investigation on DARS-AS1/miR-188-5p led to the discovery of KLF12 as a downstream target of miR-188-5p, and the regulatory pathway was established as DARS-AS1/miR-188-5p/KLF12. According to western blot results, DARS-AS1 promoted LUAD cell growth, migration, and invasion via stimulation of the PI3K/AKT pathway, activating the EMT process, and up-regulating the CyclinD1 and Bcl-2 proteins. This was the first report on the DARS-AS1/miR-188-5p/KLF12 axis and offered a novel strategy for early diagnosis, a new therapeutic method, and an improved prognosis for LUAD.


Assuntos
Adenocarcinoma de Pulmão , Fatores de Transcrição Kruppel-Like/genética , Neoplasias Pulmonares , MicroRNAs/genética , RNA Longo não Codificante/genética , Adenocarcinoma de Pulmão/genética , Adenocarcinoma de Pulmão/metabolismo , Adenocarcinoma de Pulmão/mortalidade , Adenocarcinoma de Pulmão/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Fatores de Transcrição Kruppel-Like/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/patologia , MicroRNAs/metabolismo , Invasividade Neoplásica/genética , RNA Longo não Codificante/metabolismo
16.
Biomater Sci ; 9(13): 4639-4647, 2021 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-34036971

RESUMO

Cytosolic protein delivery holds great potential for the development of protein-based biotechnologies and therapeutics. Currently, cytosolic protein delivery is mainly achieved with the assistance of various carriers. Herein, we present a universal and effective strategy for carrier-free cytosolic protein delivery via metabolic glycoengineering and bioorthogonal click reactions. Ac4ManNAz (AAM), an azido-modified N-acetylmannosamine analogue, was first employed to label tumor cell surfaces with abundant azido groups via glycometabolism. Then, proteins including RNase A, cytochrome C (Cyt C), and bovine serum albumin (BSA) were covalently modified with dibenzocyclooctyne (DBCO). Based on the highly efficient bioorthogonal click reactions between DBCO and azido, DBCO-modified proteins could be efficiently internalized by azido-labeled cancer cells. RNase A-DBCO could largely maintain its enzymatic activity and, thus, led to notable anti-tumor efficacy in HeLa and B16F10 cells in vitro and in B16F10 xenograft tumors in vivo. This study therefore provides a simple and powerful approach for carrier-free protein delivery and would have broad applicability in anti-tumor protein therapy.


Assuntos
Química Click , Neoplasias , Humanos , Neoplasias/tratamento farmacológico
18.
Biomater Sci ; 8(14): 3856-3870, 2020 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-32572407

RESUMO

siRNA-mediated RNA interference (RNAi) against inflammation-related genes provides a promising modality for the treatment of myocardial ischemia reperfusion (IR) injury, and its success is critically dependent on the development of efficient yet safe siRNA delivery vehicles. Herein, we developed a bioreducible, branched poly(ß-amino ester) with built-in redox-responsive domains (BPAE-SS) for the effective ICAM-1 siRNA delivery into injured rat cardiac microvascular endothelial cells (RCMECs). The branched BPAE-SS with a multivalent structure afforded potent siRNA binding affinity compared to its linear analogue, while upon internalization into RCMECs it was instantaneously degraded by intracellular glutathione (GSH) into small segments to mediate "on-demand" siRNA release and diminish the toxicity of post-transfection materials. By synchronizingly overcoming these critical barriers, BPAE-SS mediated remarkable ICAM-1 knockdown in IR-injured rats at 400 µg siRNA per kg via single i.v. injection, and subsequently suppressed myocardial inflammation, apoptosis, and fibrosis to recover the cardiac function. This study therefore provides a unique delivery system that can address the multiple critical challenges against non-viral siRNA delivery, and the potent therapeutic efficacy of BPAE-SS-mediated ICAM-1 silencing provides a promising strategy for the anti-inflammatory treatment of myocardial IR injury.


Assuntos
Traumatismo por Reperfusão Miocárdica , Animais , Anti-Inflamatórios , Células Endoteliais , Ésteres , Molécula 1 de Adesão Intercelular/genética , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/terapia , Polímeros , RNA Interferente Pequeno/genética , Ratos
19.
J Thorac Dis ; 12(3): 696-704, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32274135

RESUMO

BACKGROUND: The radial artery (RA) is increasingly being used for coronary artery bypass grafting (CABG). Endoscopic thoracic sympathectomy (ETS) has been shown to block innervation of sympathetic nerves of upper limbs, which reduces sweating of hands and dilates blood vessels. The modified Allen's test (MAT) is one of the commonest methods of assessing collateral arm flow prior to RA harvest, though it has limitations. However, the reliability of MAT after ETS remains unclear. We therefore investigated the effects of ETS on the results of MAT. METHODS: A retrospective cohort study was conducted on 164 consecutive Chinese patients with palmar hyperhidrosis who underwent ETS between January 2016 and January 2019. The medical records were reviewed concerning the ultrasound examination and MAT results of their RAs and ulnar arteries (UAs) in both forearms before and after ETS. RESULTS: The performance of ETS significantly increased the diameter of the right RA from 2.731±0.122 to 3.102±0.114 mm in men and from 2.347±0.074 to 2.915±0.162 mm in women. Similar effects of ETS were observed in expanding the diameters of the left RA and the UA. Meanwhile, there was no significant effect of ETS on systolic blood pressure (BP) and heart rate (HR). Overall, retesting of patients following ETS with a preoperative positive MAT result revealed a transition to a negative result. CONCLUSIONS: ETS was effective in dilating RA in both men and women, which could lead to a false negative preoperative MAT result. Patients should be questioned about their history of ETS if their RAs are to be harvested for CABG. More studies are warranted to evaluate the safety of RA as a coronary artery graft after ETS.

20.
Nano Lett ; 20(3): 1738-1746, 2020 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-32039603

RESUMO

The mucus layer and cell membrane are two major barriers against pulmonary siRNA delivery. Commonly used polycationic gene vectors can hardly penetrate the mucus layer due to the adsorption of mucin glycoproteins that trap and destabilize the polyplexes. Herein, guanidinated and fluorinated bifunctional helical polypeptides were developed to synchronizingly overcome these two barriers. The guanidine domain and α-helix facilitated trans-membrane siRNA delivery into macrophages, whereas fluorination of the polypeptides dramatically enhanced the mucus permeation capability by ∼240 folds, because incorporated fluorocarbon segments prevented adsorption of mucin glycoproteins onto polyplexes surfaces. Thus, when delivering TNF-α siRNA intratracheally, the top-performing polypeptide P7F7 provoked highly efficient gene knockdown by ∼96% at 200 µg/kg siRNA and exerted pronounced anti-inflammatory effect against acute lung injury. This study thus provides an effective strategy for transmucosal gene delivery, and it also renders promising utilities for the noninvasive, localized treatment of inflammatory pulmonary diseases.


Assuntos
Lesão Pulmonar Aguda , Macrófagos/metabolismo , Muco/metabolismo , Peptídeos , RNA Interferente Pequeno , Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/patologia , Animais , Sistemas de Liberação de Medicamentos , Técnicas de Silenciamento de Genes , Halogenação , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Peptídeos/química , Peptídeos/farmacologia , Conformação Proteica em alfa-Hélice , Células RAW 264.7 , RNA Interferente Pequeno/química , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA