Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
FASEB J ; 37(2): e22778, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36688823

RESUMEN

In Alzheimer's disease (AD) brain, inflammatory activation regulates protein levels of amyloid-ß-peptide (Aß) and phosphorylated tau (p-tau), as well as neurodegeneration; however, the regulatory mechanisms remain unclear. We constructed APP- and tau-transgenic AD mice with deletion of IKKß specifically in neurons, and observed that IKKß deficiency reduced cerebral Aß and p-tau, and modified inflammatory activation in both AD mice. However, neuronal deficiency of IKKß decreased apoptosis and maintained synaptic proteins (e.g., PSD-95 and Munc18-1) in the brain and improved cognitive function only in APP-transgenic mice, but not in tau-transgenic mice. Additionally, IKKß deficiency decreased BACE1 protein and activity in APP-transgenic mouse brain and cultured SH-SY5Y cells. IKKß deficiency increased expression of PP2A catalytic subunit isoform A, an enzyme dephosphorylating cerebral p-tau, in the brain of tau-transgenic mice. Interestingly, deficiency of IKKß in neurons enhanced autophagy as indicated by the increased ratio of LC3B-II/I in brains of both APP- and tau-transgenic mice. Thus, IKKß deficiency in neurons ameliorates AD-associated pathology in APP- and tau-transgenic mice, perhaps by decreasing Aß production, increasing p-tau dephosphorylation, and promoting autophagy-mediated degradation of BACE1 and p-tau aggregates in the brain. However, IKKß deficiency differently protects neurons in APP- and tau-transgenic mice. Further studies are needed, particularly in the context of interaction between Aß and p-tau, before IKKß/NF-κB can be targeted for AD therapies.


Asunto(s)
Enfermedad de Alzheimer , Neuroblastoma , Humanos , Ratones , Animales , Enfermedad de Alzheimer/metabolismo , Ratones Transgénicos , Quinasa I-kappa B , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Proteínas tau/metabolismo , Péptidos beta-Amiloides/metabolismo , Neuronas/metabolismo , Modelos Animales de Enfermedad
2.
Respir Res ; 24(1): 140, 2023 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-37259101

RESUMEN

BACKGROUND AND AIMS: This study aimed to investigate the expression of plasma versican and plasma exosomal versican in non-small cell lung cancer (NSCLC) and its correlation with clinicopathological features, and to evaluate its diagnostic performance in NSCLC and its predictive function for NSCLC incidence and metastasis risk. MATERIALS AND METHODS: There were 110 instances of NSCLC, 42 cases of benign lung disease, and 55 healthy controls from September 2018 to October 2020 at Tongji Hospital Affiliated to Tongji University. Blood was collected and plasma was separated before surgery, and plasma exosomes were extracted by ExoQuick kit. Morphological and molecular phenotype identification of exosomes was performed by transmission electron microscopy, Nanosight particle tracking analysis, and western blotting. Plasma versican and plasma exosomal versican were detected in all subjects to assess their expression levels and diagnostic value in NSCLC. Clinicopathological data were collected to explore correlations between abnormal plasma versican and plasma exosomal versican expression and clinicopathological parameters. Receiver operating characteristic (ROC) curve was used to judge its diagnostic performance in NSCLC, and binary logistic regression analysis was used to predict the risk of NSCLC incidence and metastasis. RESULTS: Plasma versican and plasma exosomal versican expression in NSCLC patients was significantly upregulated and was significantly higher in T3 + T4 patients compared with T1 + T2 patients (P < 0.05); the levels of plasma versican and plasma exosomal versican were positively correlated with lymph node metastasis, distant metastases (e.g., brain, bone), and mutation(e.g., EGFR,ALK)in NSCLC patients (all P < 0.05). Furthermore, ROC curve analysis showed that plasma versican and plasma exosomal versican had higher AUC values than NSE, CYFRA21-1, and SCC, and better diagnostic performance in NSCLC patients. However, the AUC and diagnostic performances of plasma versican and plasma exosomal versican in advanced-stage NSCLC patients were not shown to be significantly better than CEA. The results of binary logistic regression analysis showed that high levels of plasma exosomal versican had higher predictive value for lung cancer incidence, while high levels of plasma versican had higher predictive value for lung cancer metastasis. CONCLUSION: Our findings showed that plasma versican and plasma exosomal versican might be potential diagnostic markers for NSCLC. High plasma exosomal versican expression can be used as a predictor of NSCLC risk and high plasma versican expression can be used as a predictor of NSCLC metastasis risk.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/metabolismo , Versicanos , Biomarcadores de Tumor/genética
3.
Glia ; 69(8): 1987-2005, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33934399

RESUMEN

Growing evidence indicates that innate immune molecules regulate microglial activation in Alzheimer's disease (AD); however, their effects on amyloid pathology and neurodegeneration remain inconclusive. Here, we conditionally deleted one allele of myd88 gene specifically in microglia in APP/PS1-transgenic mice by 6 months and analyzed AD-associated pathologies by 9 months. We observed that heterozygous deletion of myd88 gene in microglia decreased cerebral amyloid ß (Aß) load and improved cognitive function of AD mice, which was correlated with reduced number of microglia in the brain and inhibited transcription of inflammatory genes, for example, tnf-α and il-1ß, in both brain tissues and individual microglia. To investigate mechanisms underlying the pathological improvement, we observed that haploinsufficiency of MyD88 increased microglial recruitment toward Aß deposits, which might facilitate Aß clearance. Microglia with haploinsufficient expression of MyD88 also increased vasculature in the brain of APP/PS1-transgenic mice, which was associated with up-regulated transcription of osteopontin and insulin-like growth factor genes in microglia. Moreover, MyD88-haploinsufficient microglia elevated protein levels of LRP1 in cerebral capillaries of APP/PS1-transgenic mice. Cell culture experiments further showed that treatments with interleukin-1ß decreased LRP1 expression in pericytes. In summary, haploinsufficiency of MyD88 in microglia at a late disease stage attenuates pro-inflammatory activation and amyloid pathology, prevents the impairment of microvasculature and perhaps also protects LRP1-mediated Aß clearance in the brain of APP/PS1-transgenic mice, all of which improves neuronal function of AD mice.


Asunto(s)
Enfermedad de Alzheimer , Microglía , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Modelos Animales de Enfermedad , Haploinsuficiencia , Ratones , Ratones Transgénicos , Microglía/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Presenilina-1/genética , Presenilina-1/metabolismo
4.
FASEB J ; 34(9): 11900-11912, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32741018

RESUMEN

Lung cancer is one of most common malignancies worldwide. We have previously identified retinoic acid-induced gene G (Rig-G) as a tumor suppressor in not only acute promyelocytic leukemia, but also in other solid tumors. However, the clinical significance of Rig-G and the underlying mechanism(s) for its biological function in lung cancer remain largely unexplored. Herein, we first compared the expression of Rig-G between lung cancer (n = 138) and normal tissues (n = 23), from public-available data sets and our patient cohort. We further analyzed the correlation of Rig-G expression with key clinico-pathological features and survival outcomes in a multi-site clinical cohort of 300 lung cancer patients. Functional studies for Rig-G were performed in cell lines, and an animal model to support clinical findings. We found that Rig-G was frequently downregulated in lung cancer tissues and cell lines, and correlated with poor prognosis in lung cancer patients. Overexpression of Rig-G led to significantly reduced cell growth and suppressed migration in A549 and NCI-H1944 cells, accompanied by reduced epithelial-mesenchymal transition. Likewise, restoration of Rig-G in Lewis lung carcinoma cells permitted development of fewer cancer metastases versus controls in an animal model. Gene expression profiling results identified p53 pathway as a key downstream target of Rig-G, and p53 inhibition by pifithrin-α caused abrogation of tumor-suppressive effects of Rig-G in lung cancer. In conclusion, we, for the first time, have identified Rig-G as a novel and important tumor suppressor, which may serve as a potential therapeutic target for restoring p53 expression in lung cancer patients.


Asunto(s)
Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Neoplasias Pulmonares/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/biosíntesis , Células A549 , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Metástasis de la Neoplasia , Proteína p53 Supresora de Tumor/genética
5.
Mol Cancer ; 19(1): 117, 2020 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-32713345

RESUMEN

BACKGROUND: Colorectal cancer (CRC) is the leading cause of cancer-related death worldwide. Exosome shave emerged as crucial regulators of intercellular communication and that abundant Circular RNAs (circRNAs) are enriched within exosomes. CircRNAs are novel members of noncoding RNAs regulating cancer proliferation and progression. However, the function and regulatory mechanism of cancer-derived exosomal circRNAs in CRC remains unclear. METHODS: CRC cells-derived exosomes were characterized using transmission electron microscopy, nanoparticle tracking analysis (NTA) and western blot. CCK-8, wound healing and transwell assays, and flow cytometry assays were conducted to assess whether exosomes would affect the proliferation, metastasis, and apoptosis of CRC cells, respectively. Moreover, we performed the RNA sequencing and RT-qPCR to identify circRNAs in exosome-stimulated CRC cells. Fluorescence in situ hybridization (FISH) assay was used to detect the cellular distribution of circPACRGL. Bioinformatic analyses (StarBase 2.0) were used to pool the miRNA targets of circPACRGL. Luciferase assays were performed to verify the direct interaction. Finally, flow cytometry was used to detect the differentiation of N1-N2 neutrophils. RESULTS: Our study identified a novel CRC-derived exosomal circRNA, circPACRGL. We found circPACRGL was significantly upregulated in CRC cells after tumor-derived exosomes addition. Moreover, circPACRGL serves as a sponge for miR-142-3p/miR-506-3p to facilitate the transforming growth factor-ß1 (TGF-ß1) expression. As a result, circPACRGL promoted CRC cell proliferation, migration and invasion, as well as differentiation of N1 to N2 neutrophils via miR-142-3p/miR-506-3p-TGF-ß1 axis. CONCLUSION: Our study, the first to reveal that cancer-derived exosomal circPACRGL plays an oncogenic role in CRC proliferation and metastasis, providing mechanistic insights into the roles of circRNAs in CRC progression and a valuable marker for CRC treatment.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Exosomas/metabolismo , MicroARNs/genética , ARN Circular/genética , Animales , Apoptosis , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Neoplasias Colorrectales/patología , Modelos Animales de Enfermedad , Exosomas/ultraestructura , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Modelos Biológicos , Interferencia de ARN , Transducción de Señal , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Microambiente Tumoral/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Acta Pharmacol Sin ; 40(2): 170-179, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29904091

RESUMEN

Although naloxone has been documented to exert neuroprotection in animal model of cerebral ischemia, the mechanism is not well understood. In this present study we investigated whether naloxone affected the mitochondrial apoptotic pathway in ischemic brain injury of rats. SD rats were subjected to a permanent middle cerebral artery occlusion surgery, and received naloxone (0.5, 1, 2 mg/kg, i.v.) immediately after ischemia. Neurological deficits were evaluated 24 h after ischemia using the McGraw Stroke Index, and then the rats were killed, and the brains were collected for further analyses. We show that naloxone treatment dose-dependently decreased the infarction volume and morphological injury, improved motor behavioral function, and markedly curtailed brain edema. Furthermore, naloxone administration significantly inhibited the nuclear translocation of NF-κB p65 and decreased the levels of nuclear NF-κB p65 in the ischemic penumbra. Naloxone administration also dose-dependently increased the NF-κB inhibitory protein (IκBα) levels and attenuated phosphorylated NIK and IKKα levels in the ischemic penumbra. In addition, naloxone administration dose-dependently increased Bcl-2 levels, decreased Bax levels, stabilized the mitochondrial transmembrane potential, and inhibited cytochrome c release and caspase 3 and caspase 9 activation. These results indicate that the neuroprotective effects of naloxone against ischemic brain injury involve the inhibition of NF-κB activation via the suppression of the NIK/IKKα/IκBα pathway and the obstruction of the mitochondrial apoptotic pathway in neurons.


Asunto(s)
Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Naloxona/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Transducción de Señal/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Quinasa I-kappa B/metabolismo , Masculino , Mitocondrias/efectos de los fármacos , FN-kappa B/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas Sprague-Dawley , Quinasa de Factor Nuclear kappa B
7.
Zhonghua Zhong Liu Za Zhi ; 37(10): 725-30, 2015 Oct.
Artículo en Zh | MEDLINE | ID: mdl-26813589

RESUMEN

OBJECTIVE: To investigate the effect of antibacterial peptide hCAP18/LL-37 on ovarian cancer microenvironment and the regulatory mechanism of its expression. METHODS: We assessed the effect of macrophage-promoted ovarian cancer cells invasion using BioCoat Matrigel invasion chamber. The expressions of hCAP18/LL-37 and versican V1 were determined by real-time PCR and Western blot analysis. SKOV3 cells were transfected with shRNA plasmid to abrogate the expression of versican V1, and then the expression of hCAP18/LL-37 in macrophages and the invasiveness of SKOV3 cells were assayed. RESULTS: The Matrigel invasion assay showed that after co-culture with macrophages for 4 days, the number of penetrated SKOV3 cells was 112.8±17.1/per high power field, significantly higher than that in the SKOV3 cells cultured alone (8.2±1.9/per high power field) (P<0.05). Addition of hCAP/LL-37 neutralizing antibody into the co-cultured macrophage-SKOV3 cells markedly inhibited the macrophage-promoted SKOV3 cells invasion. The penetrated SKOV3 cells was 22.2±5.6/per high power field, significantly lower than the 100.6±25.2/per high power field in the control macrophage- SKOV3 co-cultured cells (P<0.05). The expressions of hCAP18/LL-37 mRNA and protein in macrophages were remarkably enhanced upon co-culture with SKOV3 cells, but not changed in SKOV3 cells cultured alone. The expression and secretion of versican V1 in the ovarian cancer cells were also significantly increased after co-cultured with macrophages. Knockdown of versican V1 in SKOV3 cells by small interfering RNA significantly reduced the expression of hCAP18/LL-37 mRNA and protein in the macrophages, as well as decreased the invasiveness of SKOV3 cells (P<0.05). CONCLUSIONS: In the cancer microenvironment, the macrophage-secreted hCAP18/LL-37 promote the invasiveness of ovarian cancer cells, and the hCAP18/LL-37 expression is regulated by versican V1 protein released by ovarian cancer cells.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/farmacología , Proteínas de Neoplasias/metabolismo , Neoplasias Ováricas/patología , Microambiente Tumoral/efectos de los fármacos , Versicanos/metabolismo , Péptidos Catiónicos Antimicrobianos/metabolismo , Línea Celular Tumoral , Técnicas de Cocultivo , Colágeno , Combinación de Medicamentos , Femenino , Humanos , Laminina , Macrófagos/metabolismo , Invasividad Neoplásica , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/fisiopatología , Plásmidos , Proteoglicanos , ARN Mensajero/metabolismo , ARN Interferente Pequeño , Reacción en Cadena en Tiempo Real de la Polimerasa , Transfección , Catelicidinas
8.
Zhonghua Yi Xue Za Zhi ; 95(38): 3142-6, 2015 Oct 13.
Artículo en Zh | MEDLINE | ID: mdl-26814107

RESUMEN

OBJECTIVE: To investigate the effect of antimicrobial peptide cathelicidin secreted by non-tumorous cells in lung tumor growth. METHODS: CRAMP(-/-) mice and WT mice were used to establish a lung cancer model via tail vein injection of Lewis lung carcinoma cells (LLC1). Lung was weighted and tumor number on the lung surface was counted. Kaplan-Meier (K-M) survival curve was used to analyze survival rate of mice. Expression of cathelicidin, Ki-67 and CD68 in the tumor tissue was measured by immunohistochemical analysis. BALF cells were stained with Diff Quik and percentages of leukocyte types were determined by light microscopy. RESULTS: Cathelicidin was high expression in inflammatory cells of tumor tissue, whereas weak expression in tumor cells. The lung weight and number of tumor in CRAMP-/- mice were (0.25±0.04)g and (9.60± 2.25), respectively, which were significantly lower than those of WT mice (0.65±0.05) g and (23.40± 2.68). The difference was statistically significant (t=6.07, 3.95, all P<0.05). And Kaplan-Meier survival analysis showed median survival time of CRAMP-/- mice was 49(46-51)d, which was longer than 34(28-39) d of WT mice (χ2=12.00, P<0.05). And the positive rate of Ki-67 tumor cells was significant reduced from (35.80±2.96)% in WT mice to (18.80±2.38)% in CRAMP-/- groups (t=4.48, P<0.05). The total cell number as well as the number of lymphocytes, neutrophils, and macrophages in BALFs of CRAMP-/- mice were (4.72±0.86)×10(4), (0.08±0.02)×10(4), (0.05±0.02)×10(4) and (4.60±0.84)×10(4), respectively, while of WT mice were (16.18±1.61)×10(4), (0.32±0.05)×10(4), (0.20±0.05)×10(4) and (15.66±1.57)×10(4). All of them had significant difference (t=6.28, 4.39, 3.00, 6.20, all P<0.05). In addition, the infiltration of macrophages into lung tumors was decreased in CRAMP-/- mice compared to WT mice, from (15.53±2.28)/high power field to (6.77±3.12)/high power field (t=3.41, P<0.05). CONCLUSIONS: Non-tumor cells secreted cathelicidin promotes tumor cell proliferation and lung tumor growth. Recruitment of inflammatory cells such as macrophages into the tumor microenvironment may be the main mechanism of action.


Asunto(s)
Neoplasias Pulmonares , Animales , Antiinfecciosos , Péptidos Catiónicos Antimicrobianos , Carcinoma Pulmonar de Lewis , Catelicidinas , Progresión de la Enfermedad , Estimación de Kaplan-Meier , Pulmón , Macrófagos , Ratones , Neutrófilos
9.
J Neurosci Res ; 92(2): 232-42, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24327346

RESUMEN

We have previously reported that ginkgolides containing ginkgolides A and B (GKAB) reduce infarct size in a rat model of focal ischemia. c-Jun N-terminal kinase (JNK), also known as stress-activated kinase (SAPK), is a critical stress-responsive kinase activated by various brain insults. Previous studies have demonstrated a brief increase in p-SAPK/JNK levels after focal ischemic brain injuries. In this study, we sought to investigate whether the neuroprotective effects of GKAB in rat models of permanent focal cerebral ischemia are associated with the JNK signaling pathway. Sprague-Dawley rats were subjected to permanent middle cerebral artery occlusion by intraluminal suture blockade. GKAB was injected intravenously immediately after ischemia onset. Here we demonstrate in rats that GKAB reduces neuronal apoptosis and blocks the increase of p-SAPK/JNK levels and nuclear translocation after cerebral ischemia in a dose-dependent manner. Furthermore, we report that cerebral ischemia increases ischemia-induced induction of reactive oxygen species, and this effect was blocked by GKAB. In addition, we show that BimL is induced and attenuated by GKAB. GKAB also repressed the ischemia-induced increase in the expression of Bax and reversed the decline in expression of Bcl-2. Likewise, there was a reduction in the release or activation of several mitochondrial proapoptotic molecules, including cytochrome c, caspases 3 and 9, and PARP. Taken together, our findings strongly suggest that GKAB-mediated neuroprotective effects against focal ischemia act through the inhibition of p-SAPK/JNK activation, in which the obstruction of the mitochondrial apoptotic pathway via the JNK signaling pathway is a key downstream mechanism of GKAB.


Asunto(s)
Apoptosis/efectos de los fármacos , Isquemia Encefálica/metabolismo , Ginkgólidos/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Animales , Western Blotting , Isquemia Encefálica/patología , Modelos Animales de Enfermedad , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Lactonas/farmacología , Masculino , Mitocondrias , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno
10.
Zhonghua Zhong Liu Za Zhi ; 36(6): 412-7, 2014 Jun.
Artículo en Zh | MEDLINE | ID: mdl-25241781

RESUMEN

OBJECTIVE: The aim of this study was to investigate the mechanism of cigarette smoking (CS)-induced lung cancer growth in mice. METHODS: RelA/p65⁻/⁻ mice and WT mice were used to establish mouse models of lung cancer. Both mice were divided into two groups: air group and CS group, respectively. Tumor number on the lung surface was counted and maximal tumor size was evaluated using HE staining. Kaplan Meier (K-M) survival curve was used to analyze the survival rate of the mice. Expression of Ki-67, TNF-α and CD68 in the tumor tissue was determined by immunohistochemical analysis, and cyclin D1 and c-myc proteins were examined by Western blot. Apoptosis of tumor cells was analyzed using TUNEL staining. The concentrations of inflammatory cytokines TNF-α, IL-6 and KC in the mouse lung tissues were evaluated by ELISA. RESULTS: Compared with the WT air group, the lung weight, lung tumor multiplicity, as well as maximum tumor size in the WT mice exposed to CS were (1.5 ± 0.1)g, (64.8 ± 4.1) and (7.6 ± 0.2) mm, respectively, significantly increased than those in the WT mice not exposed to CS (P < 0.05 for all). However, there were no statistically significant differences between RelA/p65⁻/⁻ mice before and after CS exposure (P > 0.05 for all). Kaplan-Meier survival analysis showed that CS exposure significantly shortened the life time of WT mice (P < 0.05), and deletion of RelA/p65 in myeloid cells resulted in an increased survival compared with that of the WT mice (P < 0.05 for all). The ratios of Ki-67 positive tumor cells were (43.4 ± 2.9)%, (60.6 ± 5.4)%, (12.8 ± 3.6)% and (15.0 ± 4.2)% in the WT air group, WT CS groups, RelA/p65⁻/⁻ air groups and RelA/p65⁻/⁻ CS groups, respectively. After smoking, the number of Ki-67-positive cells was significantly increased in the WT mice (P < 0.05). However, there was no significant difference between the RelA/p65⁻/⁻ groups before and after smoking (P > 0.05). The apoptosis rate of WT air, WT CS, RelA/p65⁻/⁻ air and RelA/p65⁻/⁻ CS groups were (11.6 ± 1.7)%, (13.0 ± 2.0)%, (13.2 ± 2.0)% and (11.0 ± 1.4)%, respectively, with no significant difference among them (P > 0.05). Expression of cyclin D1 and c-myc was induced in response to CS exposure in lung tumor cells of WT mice. In contrast, their expressions were not significantly changed in the RelA/p65⁻/⁻ mice after smoke exposure. CS exposure was associated with an increased number of macrophages infiltrating in the tumor tissue, in both WT and RelA/p65⁻/⁻ mice (P < 0.05). The concentrations of IL-6, KC and TNF-α were significantly increased after CS exposure in the lungs of WT mice (P < 0.05). CONCLUSIONS: Cigarette smoking promotes the lung cancer growth in mice. Myeloid cell RelA/p65 mediates CS-induced tumor growth. TNFα regulated by RelA/p65 may be involved in the lung cancer development.


Asunto(s)
Neoplasias Pulmonares/inducido químicamente , Factor de Transcripción ReIA/metabolismo , Animales , Citocinas , Interleucina-6/metabolismo , Pulmón/metabolismo , Macrófagos , Masculino , Ratones , Células Mieloides/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Fumar/efectos adversos , Factor de Necrosis Tumoral alfa/metabolismo
11.
Gut Microbes ; 16(1): 2363014, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38904096

RESUMEN

Gut bacteria regulate brain pathology of Alzheimer's disease (AD) patients and animal models; however, the underlying mechanism remains unclear. In this study, 3-month-old APP-transgenic female mice with and without knock-out of Il-17a gene were treated with antibiotics-supplemented or normal drinking water for 2 months. The antibiotic treatment eradicated almost all intestinal bacteria, which led to a reduction in Il-17a-expressing CD4-positive T lymphocytes in the spleen and gut, and to a decrease in bacterial DNA in brain tissue. Depletion of gut bacteria inhibited inflammatory activation in both brain tissue and microglia, lowered cerebral Aß levels, and promoted transcription of Arc gene in the brain of APP-transgenic mice, all of which effects were abolished by deficiency of Il-17a. As possible mechanisms regulating Aß pathology, depletion of gut bacteria inhibited ß-secretase activity and increased the expression of Abcb1 and Lrp1 in the brain or at the blood-brain barrier, which were also reversed by the absence of Il-17a. Interestingly, a crossbreeding experiment between APP-transgenic mice and Il-17a knockout mice further showed that deficiency of Il-17a had already increased Abcb1 and Lrp1 expression at the blood-brain barrier. Thus, depletion of gut bacteria attenuates inflammatory activation and amyloid pathology in APP-transgenic mice via Il-17a-involved signaling pathways. Our study contributes to a better understanding of the gut-brain axis in AD pathophysiology and highlights the therapeutic potential of Il-17a inhibition or specific depletion of gut bacteria that stimulate the development of Il-17a-expressing T cells.


Asunto(s)
Enfermedad de Alzheimer , Encéfalo , Modelos Animales de Enfermedad , Microbioma Gastrointestinal , Interleucina-17 , Ratones Transgénicos , Animales , Enfermedad de Alzheimer/microbiología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Interleucina-17/metabolismo , Interleucina-17/genética , Ratones , Encéfalo/patología , Encéfalo/metabolismo , Femenino , Ratones Noqueados , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Secretasas de la Proteína Precursora del Amiloide/genética , Péptidos beta-Amiloides/metabolismo , Antibacterianos/farmacología , Ratones Endogámicos C57BL , Microglía/metabolismo , Microglía/patología , Microglía/microbiología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/microbiología , Humanos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad
12.
Clin Chim Acta ; 562: 119855, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38981565

RESUMEN

BACKGROUND AND AIMS: Bladder cancer (BCa) is a highly aggressive malignancy of the urinary system. Timely detection is imperative for enhancing BCa patient prognosis. MATERIALS AND METHODS: This study introduces a novel approach for detecting long non-coding RNA (lncRNA) Mitochondrial RNA Processing Endoribonuclease (RMRP) in urine exosomes from BCa patients using the reverse transcription recombinase-aided amplification (RT-RAA) and clustered regularly interspaced short palindromic repeats and associated Cas12a proteins (CRISPR/Cas12a) technique. Various statistical methods were used to evaluate its diagnostic value for BCa. RESULTS: The specificity of urine exosomal RMRP detection for BCa diagnosis was enhanced by using RT-RAA combined with CRISPR/Cas12a. The testing process duration was reduced to 30 min, which supports rapid detection. Moreover, this approach allows the identification of target signals in real-time using blue light, facilitating immediate detection. In clinical sample analysis, this methodology exhibited a high level of diagnostic efficacy. This was evidenced by larger area under the curve values with receiver operating characteristic curve analysis compared with using traditional RT-qPCR methods, indicating superior diagnostic accuracy and sensitivity. Furthermore, the combined analysis of RMRP expression in urine exosomes detected by RT-RAA-CRISPR/Cas12a and NMP-22 expression may further enhance diagnostic accuracy. CONCLUSIONS: The RT-RAA-CRISPR/Cas12a technology is a swift, sensitive, and uncomplicated method for nucleic acid detection. Because of its convenient and non-invasive sampling approach, user-friendly operation, and reproducibility, this technology is very promising for automated detection and holds favorable application possibilities within clinical environments.


Asunto(s)
Sistemas CRISPR-Cas , Exosomas , ARN Largo no Codificante , Neoplasias de la Vejiga Urinaria , Humanos , Neoplasias de la Vejiga Urinaria/diagnóstico , Neoplasias de la Vejiga Urinaria/orina , Neoplasias de la Vejiga Urinaria/genética , ARN Largo no Codificante/orina , ARN Largo no Codificante/genética , Exosomas/genética , Sistemas CRISPR-Cas/genética , Masculino , Persona de Mediana Edad , Femenino , Anciano
14.
Int Immunopharmacol ; 107: 108629, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35344811

RESUMEN

Mounting evidence has postulated estrogen as a contributor for lung cancer development and progression. Here, we focused on the effect of estradiol (E2) on the immune escape of non-small cell lung cancer (NSCLC). The expression of FOXO3a in NSCLC samples was screened by gene microarray and then verified using Western blot analysis in NSCLC cell lines. Interaction between E2, SIRT1, FOXO3a and PD-L1 was determined. Following ectopic expression and depletion experiments in A549 and H1435 cells, cell proliferation and killing of cytotoxic T lymphocytes (CTLs) on NSCLC cells were evaluated. Xenograft mouse models were prepared to validate the in vivo effect of E2. E2 activated SIRT1 by up-regulating the expression of ERß and thereby weakened the killing of CTLs on NSCLC cells. E2 elevated PD-L1 by up-regulating the ERß/SIRT1 axis to promote the immune escape of NSCLC cells. SIRT1 degraded FOXO3a by reducing the acetylation level of FOXO3a and increased its ubiquitination. E2 inhibited the expression of FOXO3a and elevated PD-L1 expression, thereby promoting the immune escape of NSCLC cells. In vivo results showed that E2 facilitated the growth and metastasis of NSCLC cells in nude mice by elevating ERß via SIRT1/FOXO3a/PD-L1 axis. In summary, our data revealed the critical roles of E2/ERß/SIRT1/FOXO3a/PD-L1 axis in the immune escape of NSCLC cells and suggested that the axis may be promising therapeutic targets for NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Animales , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Línea Celular Tumoral , Estradiol/farmacología , Estradiol/uso terapéutico , Receptor beta de Estrógeno/genética , Receptor beta de Estrógeno/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Ratones , Ratones Desnudos , Sirtuina 1/genética , Sirtuina 1/metabolismo
15.
J Oncol ; 2022: 2164897, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36405249

RESUMEN

Lung cancer has a complex etiology involving multiple regulatory systems. Uncertainty about the biology and evolution of lung cancer has made it difficult to improve its poor prognosis. To create efficient therapeutic targets and optimal molecular screening tools for lung cancer, the most important task seems to be to understand how it develops and progresses. The expression and regulation of GTPBP4 in non-small cell lung cancer (NSCLC) are not well understood. Using methods such as knocking down GTPBP4 in lung cancer cells and establishing a mouse lung cancer model, we found that the expression of GTPBP4 was upregulated in human lung adenocarcinoma cells and tissues, and that knocking down the expression of the GTPBP4 gene in A549 and Calu-1 lung adenocarcinoma cells can inhibit the proliferation of lung adenocarcinoma cells and reduce their invasion ability. The results of the mouse lung cancer model showed that the lung weight and the number of lung surface nodules decreased significantly in the LLC-GTPBP4 KO group. The mechanism by which GTPBP4 regulation affects the progression of lung adenocarcinoma may be related to the regulation of EMT. From this study, new research ideas emerge to explore GTPBP4 as a biomarker and therapeutic target for early diagnosis and treatment of lung cancer.

16.
Front Public Health ; 10: 850191, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35387184

RESUMEN

Objective: To investigate whether first-trimester fasting plasma glucose (FPG), blood coagulation function and lipid metabolism could predict gestational diabetes mellitus (GDM) risk. Methods: From October 2020 to May 2021, a total of 584 pregnant women who took prenatal care in Shanghai Jiaotong University Affiliated Sixth People's Hospital were chosen as the observation subjects. The clinical information and serum samples of all pregnant women were collected at 10-13 weeks of gestation and the blood coagulation function, fasting blood glucose and lipid profiles of the pregnant women were detected. A 75 g oral glucose tolerance test was performed up to 24-28 weeks of gestation. One hundred forty-two pregnant women with GDM and 442 pregnant women without GDM were detected. Data were expressed by x ± s or median (interquartile range) and were analyzed using student's t-test, Wilcoxon rank sum test and Logistic regression analysis. The area under the curve (AUC) was calculated by receiver operating characteristic curve (ROC) to analyze the predictive values. Results: Compared with non-GDM group, age, pre-pregnancy BMI, FPG, FIB, D-Dimer, FDP, FPG, TC, TG, LDL-C, sdLDL-C, APOB and APOE in GDM group were significantly higher than those in non-GDM group, while PT, INR, APTT and TT were significantly lower than those in non-GDM group. Univariate logistic regression analysis was used to explore the risk factors of GDM. Gestational age, pre-pregnancy BMI, FPG, PT, INR, APTT, FIB, TT, D-Dimer, TC, TG, LDL-C, sdLDL-C, APOB and APOE were all independent predictors of GDM. Multivariatelogistic regression showed that pre-pregnancy BMI, FPG, APTT, TT, TG, LDL-C, sdLDL-C and APOB were risk factors for GDM. The AUC of the established GDM risk prediction model was 0.892 (0.858-0.927), and the sensitivity and specificity were 80.71 and 86.85%, respectively; which were greater than that of pre-pregnancy BMI, FPG, APTT, TT,TG, LDL-C, sdLDL-C, APOB alone, and the difffference was statistically signifificant (P < 0.05). Conclusions: FPG, APTT, TT, TG, LDL-C, sdLDL-C, APOB and pre-pregnancy BMI in early pregnancy has important clinical value for the prediction of GDM, We combined these laboratory indicators and established a GDM risk prediction model, which is conducive to the early identification, intervention and treatment of GDM, so as to reduce the morbidity of maternal and infant complications.


Asunto(s)
Diabetes Gestacional , Apolipoproteínas B/metabolismo , Apolipoproteínas E/metabolismo , Coagulación Sanguínea , Glucemia/análisis , Glucemia/metabolismo , Índice de Masa Corporal , LDL-Colesterol/metabolismo , Diabetes Gestacional/diagnóstico , Diagnóstico Precoz , Femenino , Glucolípidos , Humanos , Metabolismo de los Lípidos , Embarazo
17.
J Immunol Res ; 2022: 9444227, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35664352

RESUMEN

Asthma is a heterogeneous inflammatory disorder of the airways, and multiple studies have addressed the vital role of the nucleotide-binding oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3)/caspase-1/interleukin-1ß (IL-1ß) pathway in asthma, but its impact on ovalbumin- (OVA-) induced neutrophilic asthma remains unclear. Here, we explored this pathway's effect on airway inflammation in neutrophilic asthma to clarify whether blocking this signaling could alleviate asthmatic airway inflammation. Using an established OVA-induced neutrophilic asthma mouse model, we provided asthmatic mice with a highly selective NLRP3 inhibitor, MCC950, and a specific caspase-1 inhibitor, Ac-YVAD-cmk. Our results indicated that asthmatic mice exhibited increased airway hyperresponsiveness, neutrophil infiltration, and airway mucus hypersecretion, upregulated retinoid-related orphan receptor-γt (RORγt) mRNA expression, and downregulated fork head box p3 (Foxp3) mRNA expression, which was concurrent with NLRP3 inflammasome activation and upregulation of caspase-1, IL-1ß, and IL-18 expression in lung. Treatment of NLRP3 inflammasome inhibitors significantly attenuated airway hyperresponsiveness, airway inflammation, and reversed T helper 17 (Th17)/regulatory T (Treg) cell imbalance in asthmatic mice. We propose that the NLRP3/caspase-1/IL-1ß pathway plays an important role in the pathological process of neutrophilic asthma and provides evidence that blocking this pathway could potentially be a treatment strategy to ameliorate airway inflammation in asthma after validation with future experimental and clinical studies.


Asunto(s)
Asma , Hipersensibilidad Respiratoria , Animales , Caspasa 1 , Modelos Animales de Enfermedad , Inflamasomas/metabolismo , Inflamación , Interleucina-1beta/metabolismo , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Ovalbúmina , ARN Mensajero , Linfocitos T Reguladores
18.
Front Oncol ; 12: 1118042, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36591456

RESUMEN

[This corrects the article DOI: 10.3389/fonc.2022.942964.].

19.
Front Oncol ; 12: 942964, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36353553

RESUMEN

The underlying molecular mechanisms and evolutionary patterns of lung cancer metastasis remain unclear, resulting in a lack of effective indicators for early diagnosis of metastasis. We retrospectively analyzed 117 patients with primary non-small cell lung cancer (NSCLC) admitted to Tongji Hospital of Tongji University in 2021, of which 93 patients with tumor metastasis were set as the metastasis group. 24 patients without metastasis were set as the non-metastasis group. The differences of each index in the two groups of patients and the expression levels in different TNM stages were compared. This study intends to evaluate the diagnostic value and net clinical benefit of common blood-related indicators Neutrophil/lymphocyte (NLR), lymphocyte/monocyte (LMR), High density lipoprotein/neutrophil (HNR), High density lipoprotein/monocyte (HMR) and combined assays in NSCLC metastasis for the early diagnosis of patients with NSCLC metastasis. It was found that the level of NLR was higher in metastatic NSCLC than non-metastatic, but the level of LMR, HNR and HMR was lower. The levels of NLR, LMR, HNR and HMR in patients with different TNM stages showed that NLR levels increased with TNM stage, while LMR, HNR and HMR levels decreased. The threshold probability range of the 4 combined tests was greater and the overall clinical benefit rate was higher compared to the individual tests. Our findings suggest that NLR, LMR, HNR and HMR have better diagnostic value for NSCLC metastasis. This study provides a clinical basis for investigating the mechanisms by which immune cells and lipid metabolism-related proteins remodel the microenvironment prior to NSCLC metastasis.

20.
J Oncol ; 2022: 5242437, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35756082

RESUMEN

Treatment of cancer in humans requires a thorough understanding of the multiple pathways by which it develops. Recent studies suggest that nuclear receptor coactivator 4 (NCOA4) may be a predictive biomarker for renal cancer. In the present work, TCGA, GEPIA, and several bioinformatics approaches were used to analyze the NCOA4 expression patterns, prognostic relevance, and association between NCOA4 and clinicopathological features and immune cell infiltration. We investigated NCOA4 expression in malignancies. Low NCOA4 expression was associated with poor overall survival in individuals with malignancies such as cholangiocarcinoma, colon adenocarcinoma, and clear cell renal carcinoma. We also analyzed NCOA4 DNA methylation in normal and primary tumor tissues and investigated possible functional pathways underlying NCOA4-mediated oncogenesis. In conclusion, downregulation of NCOA4 is associated with poor prognosis, and NCOA4 may be a predictive biomarker for COAD.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA