Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Sci Adv ; 10(28): eadl4913, 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-38985878

RESUMEN

The pathophysiology of silicosis is poorly understood, limiting development of therapies for those who have been exposed to the respirable particle. We explored mechanisms of silica-induced pulmonary fibrosis in human lung samples collected from patients with occupational exposure to silica and in a longitudinal mouse model of silicosis using multiple modalities including whole-lung single-cell RNA sequencing and histological, biochemical, and physiologic assessments. In addition to pulmonary inflammation and fibrosis, intratracheal silica challenge induced osteoclast-like differentiation of alveolar macrophages and recruited monocytes, driven by induction of the osteoclastogenic cytokine, receptor activator of nuclear factor κΒ ligand (RANKL) in pulmonary lymphocytes, and alveolar type II cells. Anti-RANKL monoclonal antibody treatment suppressed silica-induced osteoclast-like differentiation in the lung and attenuated pulmonary fibrosis. We conclude that silica induces differentiation of pulmonary osteoclast-like cells leading to progressive lung injury, likely due to sustained elaboration of bone-resorbing proteases and hydrochloric acid. Interrupting osteoclast-like differentiation may therefore constitute a promising avenue for moderating lung damage in silicosis.


Asunto(s)
Diferenciación Celular , Osteoclastos , Fibrosis Pulmonar , Dióxido de Silicio , Silicosis , Dióxido de Silicio/toxicidad , Animales , Humanos , Osteoclastos/metabolismo , Osteoclastos/efectos de los fármacos , Osteoclastos/patología , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/metabolismo , Ratones , Silicosis/patología , Silicosis/metabolismo , Silicosis/etiología , Diferenciación Celular/efectos de los fármacos , Ligando RANK/metabolismo , Modelos Animales de Enfermedad , Masculino , Pulmón/patología , Pulmón/metabolismo , Pulmón/efectos de los fármacos , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patología , Macrófagos Alveolares/efectos de los fármacos , Femenino
2.
Sci Adv ; 9(19): eadf8549, 2023 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-37163604

RESUMEN

Lymphangioleiomyomatosis (LAM) is a rare, progressive lung disease that predominantly affects women. LAM cells carry TSC1/TSC2 mutations, causing mTORC1 hyperactivation and uncontrolled cell growth. mTORC1 inhibitors stabilize lung function; however, sustained efficacy requires long-term administration, and some patients fail to tolerate or respond to therapy. Although the genetic basis of LAM is known, mechanisms underlying LAM pathogenesis remain elusive. We integrated single-cell RNA sequencing and single-nuclei ATAC-seq of LAM lungs to construct a gene regulatory network controlling the transcriptional program of LAM cells. We identified activation of uterine-specific HOX-PBX transcriptional programs in pulmonary LAMCORE cells as regulators of cell survival depending upon HOXD11-PBX1 dimerization. Accordingly, blockage of HOXD11-PBX1 dimerization by HXR9 suppressed LAM cell survival in vitro and in vivo. PBX1 regulated STAT1/3, increased the expression of antiapoptotic genes, and promoted LAM cell survival in vitro. The HOX-PBX gene network provides promising targets for treatment of LAM/TSC mTORC1-hyperactive cancers.


Asunto(s)
Redes Reguladoras de Genes , Proteínas de Homeodominio , Linfangioleiomiomatosis , Humanos , Análisis de la Célula Individual , Linfangioleiomiomatosis/metabolismo , Linfangioleiomiomatosis/patología , Factores de Transcripción/metabolismo , Pulmón/metabolismo , Pulmón/patología , Animales , Ratas , Metástasis de la Neoplasia , Multiómica , Femenino
3.
Nat Commun ; 14(1): 1205, 2023 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-36864068

RESUMEN

Pulmonary alveolar microlithiasis is an autosomal recessive lung disease caused by a deficiency in the pulmonary epithelial Npt2b sodium-phosphate co-transporter that results in accumulation of phosphate and formation of hydroxyapatite microliths in the alveolar space. The single cell transcriptomic analysis of a pulmonary alveolar microlithiasis lung explant showing a robust osteoclast gene signature in alveolar monocytes and the finding that calcium phosphate microliths contain a rich protein and lipid matrix that includes bone resorbing osteoclast enzymes and other proteins suggested a role for osteoclast-like cells in the host response to microliths. While investigating the mechanisms of microlith clearance, we found that Npt2b modulates pulmonary phosphate homeostasis through effects on alternative phosphate transporter activity and alveolar osteoprotegerin, and that microliths induce osteoclast formation and activation in a receptor activator of nuclear factor-κB ligand and dietary phosphate dependent manner. This work reveals that Npt2b and pulmonary osteoclast-like cells play key roles in pulmonary homeostasis and suggest potential new therapeutic targets for the treatment of lung disease.


Asunto(s)
Enfermedades Pulmonares , Osteogénesis , Humanos , Homeostasis , Pulmón
4.
JCI Insight ; 8(9)2023 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-36927688

RESUMEN

Tuberous sclerosis complex (TSC) is characterized by multisystem, low-grade neoplasia involving the lung, kidneys, brain, and heart. Lymphangioleiomyomatosis (LAM) is a progressive pulmonary disease affecting almost exclusively women. TSC and LAM are both caused by mutations in TSC1 and TSC2 that result in mTORC1 hyperactivation. Here, we report that single-cell RNA sequencing of LAM lungs identified activation of genes in the sphingolipid biosynthesis pathway. Accordingly, the expression of acid ceramidase (ASAH1) and dihydroceramide desaturase (DEGS1), key enzymes controlling sphingolipid and ceramide metabolism, was significantly increased in TSC2-null cells. TSC2 negatively regulated the biosynthesis of tumorigenic sphingolipids, and suppression of ASAH1 by shRNA or the inhibitor ARN14976 (17a) resulted in markedly decreased TSC2-null cell viability. In vivo, 17a significantly decreased the growth of TSC2-null cell-derived mouse xenografts and short-term lung colonization by TSC2-null cells. Combined rapamycin and 17a treatment synergistically inhibited renal cystadenoma growth in Tsc2+/- mice, consistent with increased ASAH1 expression and activity being rapamycin insensitive. Collectively, the present study identifies rapamycin-insensitive ASAH1 upregulation in TSC2-null cells and tumors and provides evidence that targeting aberrant sphingolipid biosynthesis pathways has potential therapeutic value in mechanistic target of rapamycin complex 1-hyperactive neoplasms, including TSC and LAM.


Asunto(s)
Neoplasias Pulmonares , Esclerosis Tuberosa , Humanos , Ratones , Femenino , Animales , Esclerosis Tuberosa/tratamiento farmacológico , Proteínas Supresoras de Tumor/genética , Regulación hacia Arriba , Ceramidasa Ácida/genética , Ceramidasa Ácida/metabolismo , Ceramidasa Ácida/uso terapéutico , Neoplasias Pulmonares/patología , Sirolimus/farmacología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones Noqueados
5.
bioRxiv ; 2023 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-36824953

RESUMEN

The pathophysiology of silicosis is poorly understood, limiting development of therapies for those who have been exposed to the respirable particle. We explored the mechanisms of silica-induced pulmonary fibrosis in a mouse model using multiple modalities including whole-lung single-nucleus RNA sequencing. These analyses revealed that in addition to pulmonary inflammation and fibrosis, intratracheal silica challenge induced osteoclast-like differentiation of alveolar macrophages and recruited monocytes, driven by induction of the osteoclastogenic cytokine, receptor activator of nuclear factor-κB ligand (RANKL) in pulmonary lymphocytes and alveolar type II cells. Furthermore, anti-RANKL monoclonal antibody treatment suppressed silica-induced osteoclast-like differentiation in the lung and attenuated silica-induced pulmonary fibrosis. We conclude that silica induces osteoclast-like differentiation of distinct recruited and tissue resident monocyte populations, leading to progressive lung injury, likely due to sustained elaboration of bone resorbing proteases and hydrochloric acid. Interrupting osteoclast-like differentiation may therefore constitute a promising avenue for moderating lung damage in silicosis.

6.
Am J Pathol ; 193(2): 191-200, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36336066

RESUMEN

Kidney cyst expansion in tuberous sclerosis complex (TSC) or polycystic kidney disease (PKD) requires active secretion of chloride (Cl-) into the cyst lumen. In PKD, Cl- secretion is primarily mediated via the cystic fibrosis transmembrane conductance regulator (CFTR) in principal cells. Kidney cystogenesis in TSC is predominantly composed of type A intercalated cells, which do not exhibit noticeable expression of CFTR. The identity of the Cl--secreting molecule(s) in TSC cyst epithelia remains speculative. RNA-sequencing analysis results were used to examine the expression of FOXi1, the chief regulator of acid base transporters in intercalated cells, along with localization of Cl- channel 5 (ClC5), in various models of TSC. Results from Tsc2+/- mice showed that the expansion of kidney cysts corresponded to the induction of Foxi1 and correlated with the appearance of ClC5 and H+-ATPase on the apical membrane of cyst epithelia. In various mouse models of TSC, Foxi1 was robustly induced in the kidney, and ClC5 and H+-ATPase were expressed on the apical membrane of cyst epithelia. Expression of ClC5 was also detected on the apical membrane of cyst epithelia in humans with TSC but was absent in humans with autosomal dominant PKD or in a mouse model of PKD. These results indicate that ClC5 is expressed on the apical membrane of cyst epithelia and is a likely candidate mediating Cl- secretion into the kidney cyst lumen in TSC.


Asunto(s)
Quistes , Enfermedades Renales Poliquísticas , Esclerosis Tuberosa , Humanos , Animales , Ratones , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Cloruros/metabolismo , Esclerosis Tuberosa/metabolismo , Riñón/metabolismo , Epitelio/metabolismo , Factores de Transcripción Forkhead/metabolismo
7.
Int J Mol Sci ; 23(24)2022 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-36555783

RESUMEN

The mechanistic target of rapamycin (mTOR) is a serine/threonine protein kinase that regulates multiple processes, including gene transcription, protein synthesis, ribosome biogenesis, autophagy, cell metabolism, and cell growth [...].


Asunto(s)
Transducción de Señal , Serina-Treonina Quinasas TOR , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Transducción de Señal/genética , Biosíntesis de Proteínas , Autofagia
8.
Lancet Respir Med ; 9(11): 1313-1327, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34461049

RESUMEN

Lymphangioleiomyomatosis (LAM) is a slowly progressive, low-grade, metastasising neoplasm of women, characterised by infiltration of the lung parenchyma with abnormal smooth muscle-like cells, resulting in cystic lung destruction. The invading cell in LAM arises from an unknown source and harbours mutations in tuberous sclerosis complex (TSC) genes that result in constitutive activation of the mechanistic target of rapamycin (mTOR) pathway, dysregulated cellular proliferation, and a programme of frustrated lymphangiogenesis, culminating in disordered lung remodelling and respiratory failure. Over the past two decades, all facets of LAM basic and clinical science have seen important advances, including improved understanding of molecular mechanisms, novel diagnostic and prognostic biomarkers, effective treatment strategies, and comprehensive clinical practice guidelines. Further research is needed to better understand the natural history of LAM; develop more powerful diagnostic, prognostic, and predictive biomarkers; optimise the use of inhibitors of mTOR complex 1 in the treatment of LAM; and explore novel approaches to the development of remission-inducing therapies.


Asunto(s)
Neoplasias Pulmonares , Linfangioleiomiomatosis , Femenino , Humanos , Pulmón/patología , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Linfangioleiomiomatosis/etiología , Linfangioleiomiomatosis/genética , Mutación , Sirolimus/uso terapéutico
9.
Int J Mol Sci ; 22(9)2021 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-33922083

RESUMEN

In the connectivity map (CMap) approach to drug repositioning and development, transcriptional signature of disease is constructed by differential gene expression analysis between the diseased tissue or cells and the control. The negative correlation between the transcriptional disease signature and the transcriptional signature of the drug, or a bioactive compound, is assumed to indicate its ability to "reverse" the disease process. A major limitation of traditional CMaP analysis is the use of signatures derived from bulk disease tissues. Since the key driver pathways are most likely dysregulated in only a subset of cells, the "averaged" transcriptional signatures resulting from bulk analysis lack the resolution to effectively identify effective therapeutic agents. The use of single-cell RNA-seq (scRNA-seq) transcriptomic assay facilitates construction of disease signatures that are specific to individual cell types, but methods for using scRNA-seq data in the context of CMaP analysis are lacking. Lymphangioleiomyomatosis (LAM) mutations in TSC1 or TSC2 genes result in the activation of the mTOR complex 1 (mTORC1). The mTORC1 inhibitor Sirolimus is the only FDA-approved drug to treat LAM. Novel therapies for LAM are urgently needed as the disease recurs with discontinuation of the treatment and some patients are insensitive to the drug. We developed methods for constructing disease transcriptional signatures and CMaP analysis using scRNA-seq profiling and applied them in the analysis of scRNA-seq data of lung tissue from naïve and sirolimus-treated LAM patients. New methods successfully implicated mTORC1 inhibitors, including Sirolimus, as capable of reverting the LAM transcriptional signatures. The CMaP analysis mimicking standard bulk-tissue approach failed to detect any connection between the LAM signature and mTORC1 signaling. This indicates that the precise signature derived from scRNA-seq data using our methods is the crucial difference between the success and the failure to identify effective therapeutic treatments in CMaP analysis.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Conectoma/métodos , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/patología , Linfangioleiomiomatosis/patología , Análisis de la Célula Individual/métodos , Serina-Treonina Quinasas TOR/metabolismo , Antibióticos Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/genética , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Linfangioleiomiomatosis/tratamiento farmacológico , Linfangioleiomiomatosis/genética , Linfangioleiomiomatosis/metabolismo , Pronóstico , Análisis de Secuencia de ARN , Sirolimus/uso terapéutico , Serina-Treonina Quinasas TOR/genética
10.
Proc Natl Acad Sci U S A ; 118(6)2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33536341

RESUMEN

Tuberous sclerosis complex (TSC) is caused by mutations in either TSC1 or TSC2 genes and affects multiple organs, including kidney, lung, and brain. In the kidney, TSC presents with the enlargement of benign tumors (angiomyolipomata) and cysts, which eventually leads to kidney failure. The factors promoting cyst formation and tumor growth in TSC are incompletely understood. Here, we report that mice with principal cell-specific inactivation of Tsc1 develop numerous cortical cysts, which are overwhelmingly composed of hyperproliferating A-intercalated (A-IC) cells. RNA sequencing and confirmatory expression studies demonstrated robust expression of Forkhead Transcription Factor 1 (Foxi1) and its downstream targets, apical H+-ATPase and cytoplasmic carbonic anhydrase 2 (CAII), in cyst epithelia in Tsc1 knockout (KO) mice but not in Pkd1 mutant mice. In addition, the electrogenic 2Cl-/H+ exchanger (CLC-5) is significantly up-regulated and shows remarkable colocalization with H+-ATPase on the apical membrane of cyst epithelia in Tsc1 KO mice. Deletion of Foxi1, which is vital to intercalated cells viability and H+-ATPase expression, completely abrogated the cyst burden in Tsc1 KO mice, as indicated by MRI images and histological analysis in kidneys of Foxi1/Tsc1 double-knockout (dKO) mice. Deletion of CAII, which is critical to H+-ATPase activation, caused significant reduction in cyst burden and increased life expectancy in CAII/Tsc1 dKO mice vs. Tsc1 KO mice. We propose that intercalated cells and their acid/base/electrolyte transport machinery (H+-ATPase/CAII/CLC-5) are critical to cystogenesis, and their inhibition or inactivation is associated with significant protection against cyst generation and/or enlargement in TSC.


Asunto(s)
Anhidrasa Carbónica II/genética , Factores de Transcripción Forkhead/genética , Insuficiencia Renal/genética , Proteína 1 del Complejo de la Esclerosis Tuberosa/genética , Animales , Quistes/genética , Quistes/patología , Modelos Animales de Enfermedad , Humanos , Riñón/metabolismo , Riñón/patología , Ratones , Mutación/genética , ATPasas de Translocación de Protón/genética , Insuficiencia Renal/patología , Canales Catiónicos TRPP/genética , Esclerosis Tuberosa
12.
3 Biotech ; 10(11): 485, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33117626

RESUMEN

Our study aims to investigate the impact of miR-224 on cell migration and invasion in colorectal cancer (CRC) as well as its molecular mechanisms. The results showed that miR-224 was significantly upregulated in CRC compared to normal tissues via the TCGA database. Overexpression of miR-224 promoted CRC cell migration and invasion, while inhibition of miR-224 demonstrated the opposite result via transwell assays. In addition, we found that BTRC was a target gene of miR-224 through the miRecords database and dual-luciferase assay, while western blot together with RT-qPCR showed that inhibition of miR-224 led to elevated BTRC expression in protein level but not in mRNA level, and also decreased the expression of ß-catenin. In reference to the Human Protein Atlas, BTRC protein expression was higher in normal tissues than in CRC tissues. In conclusion, miR-224 regulates its target BTRC protein expression and its related Wnt/ß-catenin pathway. Its impact on cell migration and invasion in CRC cells suggested that miR-224 could be a prospective therapeutic target for early-stage non-metastatic CRC.

13.
Orphanet J Rare Dis ; 15(1): 209, 2020 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-32807195

RESUMEN

BACKGROUND: Tuberous sclerosis complex (TSC) is a genetic disorder that cause tumors to form in many organs. These lesions may lead to epilepsy, autism, developmental delay, renal, and pulmonary failure. Loss of function mutations in TSC1 and TSC2 genes by aberrant activation of the mechanistic target of rapamycin (mTORC1) signaling pathway are the known causes of TSC. Therefore, targeting mTORC1 becomes a most available therapeutic strategy for TSC. Although mTORC1 inhibitor rapamycin and Rapalogs have demonstrated exciting results in the recent clinical trials, however, tumors rebound and upon the discontinuation of the mTORC1 inhibition. Thus, understanding the underlying molecular mechanisms responsible for rapamycin-induced cell survival becomes an urgent need. Identification of additional molecular targets and development more effective remission-inducing therapeutic strategies are necessary for TSC patients. RESULTS: We have discovered an Mitogen-activated protein kinase (MAPK)-evoked positive feedback loop that dampens the efficacy of mTORC1 inhibition. Mechanistically, mTORC1 inhibition increased MEK1-dependent activation of MAPK in TSC-deficient cells. Pharmacological inhibition of MAPK abrogated this feedback loop activation. Importantly, the combinatorial inhibition of mTORC1 and MAPK induces the death of TSC2-deficient cells. CONCLUSIONS: Our results provide a rationale for dual targeting of mTORC1 and MAPK pathways in TSC and other mTORC1 hyperactive neoplasm.


Asunto(s)
Esclerosis Tuberosa , Supervivencia Celular , Humanos , Proteínas Quinasas Activadas por Mitógenos , Sirolimus/farmacología , Sirolimus/uso terapéutico , Esclerosis Tuberosa/tratamiento farmacológico , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteína 2 del Complejo de la Esclerosis Tuberosa
14.
Am J Respir Crit Care Med ; 202(10): 1373-1387, 2020 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-32603599

RESUMEN

Rationale: Lymphangioleiomyomatosis (LAM) is a metastatic neoplasm of reproductive-age women associated with mutations in tuberous sclerosis complex genes. LAM causes cystic remodeling of the lung and progressive respiratory failure. The sources and cellular characteristics of LAM cells underlying disease pathogenesis remain elusive.Objectives: Identification and characterization of LAM cells in human lung and uterus using a single-cell approach.Methods: Single-cell and single-nuclei RNA sequencing on LAM (n = 4) and control (n = 7) lungs, immunofluorescence confocal microscopy, ELISA, and aptamer proteomics were used to identify and validate LAMCORE cells and secreted biomarkers, predict cellular origins, and define molecular and cellular networks in LAM.Measurements and Main Results: A unique cell type termed LAMCORE was identified, which was distinct from, but closely related to, lung mesenchymal cells. LAMCORE cells expressing signature genes included known LAM markers such as PMEL, FIGF, CTSK, and MLANA and novel biomarkers validated by aptamer screening, ELISA, and immunofluorescence microscopy. LAM cells in lung and uterus are morphologically indistinguishable and share similar gene expression profiles and biallelic TSC2 mutations, supporting a potential uterine origin for the LAMCORE cell. Effects of LAM on resident pulmonary cell types indicated recruitment and activation of lymphatic endothelial cells.Conclusions: A unique population of LAMCORE cells was identified in lung and uterus of patients with LAM, sharing close transcriptomic identity. LAM cell selective markers, secreted biomarkers, and the predicted cellular molecular features provide new insights into the signaling and transcriptional programs that may serve as diagnostic markers and therapeutic targets to influence the pathogenesis of LAM.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/genética , Linfangioleiomiomatosis/diagnóstico , Linfangioleiomiomatosis/genética , Transcriptoma/genética , Neoplasias Uterinas/diagnóstico , Neoplasias Uterinas/genética , Adulto , Anciano , Femenino , Humanos , Persona de Mediana Edad , Análisis de la Célula Individual , Estados Unidos
15.
PLoS One ; 15(2): e0228894, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32078667

RESUMEN

Lymphangioleiomyomatosis (LAM) is a devastating lung disease caused by inactivating gene mutations in either TSC1 or TSC2 that result in hyperactivation of the mechanistic target of rapamycin complex 1 (mTORC1). As LAM occurs predominantly in women during their reproductive age and is exacerbated by pregnancy, the female hormonal environment, and in particular estrogen, is implicated in LAM pathogenesis and progression. However, detailed underlying molecular mechanisms are not well understood. In this study, utilizing human pulmonary LAM specimens and cell culture models of TSC2-deficient LAM patient-derived and rat uterine leiomyoma-derived cells, we tested the hypothesis that estrogen promotes the growth of mTORC1-hyperactive cells through pyruvate kinase M2 (PKM2). Estrogen increased the phosphorylation of PKM2 at Ser37 and induced the nuclear translocation of phospho-PKM2. The estrogen receptor antagonist Faslodex reversed these effects. Restoration of TSC2 inhibited the phosphorylation of PKM2 in an mTORC1 inhibitor-insensitive manner. Finally, accumulation of phosphorylated PKM2 was evident in pulmonary nodule from LAM patients. Together, our data suggest that female predominance of LAM might be at least in part attributed to estrogen stimulation of PKM2-mediated cellular metabolic alterations. Targeting metabolic regulators of PKM2 might have therapeutic benefits for women with LAM and other female-specific neoplasms.


Asunto(s)
Estrógenos/metabolismo , Piruvato Quinasa/metabolismo , Proteína 2 del Complejo de la Esclerosis Tuberosa/genética , Animales , Línea Celular Tumoral , Estrógenos/fisiología , Femenino , Humanos , Pulmón/patología , Neoplasias Pulmonares/patología , Linfangioleiomiomatosis/genética , Linfangioleiomiomatosis/fisiopatología , Diana Mecanicista del Complejo 1 de la Rapamicina , Fosforilación , Piruvato Quinasa/fisiología , Ratas , Transducción de Señal/efectos de los fármacos , Proteína 1 del Complejo de la Esclerosis Tuberosa/genética , Proteína 1 del Complejo de la Esclerosis Tuberosa/metabolismo , Proteína 2 del Complejo de la Esclerosis Tuberosa/metabolismo , Proteínas Supresoras de Tumor/genética
16.
Front Oncol ; 10: 597569, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33614483

RESUMEN

BACKGROUND: Autophagy is a "self-feeding" phenomenon of cells, which is crucial in mammalian development. Long non-coding RNA (lncRNA) is a new regulatory factor for cell autophagy, which can regulate the process of autophagy to affect tumor progression. However, poor attention has been paid to the roles of autophagy-related lncRNAs in breast cancer. OBJECTIVE: This study aimed to construct an autophagy-related lncRNA signature that can effectively predict the prognosis of breast cancer patients and explore the potential functions of these lncRNAs. METHODS: The RNA sequencing (RNA-Seq) data of breast cancer patients was collected from The Cancer Genome Atlas (TCGA) database and the GSE20685 database. Multivariate Cox analysis was implemented to produce an autophagy-related lncRNA signature in the TCGA cohort. The signature was then validated in the GSE20685 cohort. The receiver operator characteristic (ROC) curve was performed to evaluate the predictive ability of the signature. Gene set enrichment analysis (GSEA) was used to explore the potential functions based on the signature. Finally, the study developed a nomogram and internal verification based on the autophagy-related lncRNAs. RESULTS: A signature composed of 9 autophagy-related lncRNAs was determined as a prognostic model, and 1,109 breast cancer patients were divided into high-risk group and low-risk group based on median risk score of the signature. Further analysis demonstrated that the over survival (OS) of breast cancer patients in the high-risk group was poorer than that in the low-risk group based on the prognostic signature. The area under the curve (AUC) of ROC curve verified the sensitivity and specificity of this signature. Additionally, we confirmed the signature is an independent factor and found it may be correlated to the progression of breast cancer. GSEA showed gene sets were notably enriched in carcinogenic activation pathways and autophagy-related pathways. The qRT-PCR identified 5 lncRNAs with significantly differential expression in breast cancer cells based on the 9 lncRNAs of the prognostic model, and the results were consistent with the tissues. CONCLUSION: In summary, our signature has potential predictive value in the prognosis of breast cancer and these autophagy-related lncRNAs may play significant roles in the diagnosis and treatment of breast cancer.

17.
Brain Sci ; 9(11)2019 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-31739561

RESUMEN

PURPOSE: Previous research has evaluated the effects of acute hypoxia exposure on cognitive function, notably executive function. No studies, to date, have evaluated the effects of acute hypoxia exposure on memory interference, which was the purpose of this experiment. METHODS: A within-subjects, counterbalanced experimental design was employed, with condition (hypoxia vs. normoxia) and time (immediate vs. delayed) being the independent variables. Participants (N = 21; Mage = 21.0 years) completed two laboratory visits, involving 30 min of exposure to either hypoxia (FIO2 = 0.12) or normoxia (FIO2 = 0.21). Following this, they completed a memory interference task (AB/AC paradigm), assessing immediate and delayed proactive and retroactive interference. RESULTS: For retroactive interference, we observed a significant main effect for condition, F(1, 20) = 5.48, p = 0.03, ƞ2 = 0.10, condition by time interaction, F(1, 20) = 4.96, p = 0.03, ƞ2 = 0.01, but no main effect for time, F(1, 20) = 1.75, p = 0.20, ƞ2 = 0.004. CONCLUSION: Our results demonstrate that acute hypoxia exposure was facilitative in reducing memory interference. We discuss these findings in the context of the potential therapeutic effects of acute hypoxia exposure on synaptic plasticity.

18.
Artículo en Inglés | MEDLINE | ID: mdl-31480233

RESUMEN

Schools are salient locations for children with disabilities to accrue physical activity (PA) and to diminish sedentary time (ST). We examined seasonal variation in accelerometer-assessed PA and ST among children with disabilities during the school day in three school settings (physical education (PE) lessons, recess and lunchtime). Children (n = 270) from 13 special schools for those with five disability types (visual impairments, hearing impairments, physical disabilities, intellectual disabilities (ID), and social development problems) participated. Their PA and ST were assessed during three winter and three summer school days using accelerometry. Linear mixed models were performed to determine seasonal variation in the proportion of time they spent in moderate-to-vigorous physical activity (MVPA) and ST in the three settings. On average, the children spent 4.5% (18.6 min) and 4.0% (15.6 min) in MVPA at school during winter and summer days, respectively. They were more physically active during winter (especially during recess and lunchtime), but there were no seasonal differences for ST. Thus, children's year-round engagement in PA needs to be promoted, especially during summer.


Asunto(s)
Acelerometría/estadística & datos numéricos , Niños con Discapacidad/estadística & datos numéricos , Ejercicio Físico , Conducta Sedentaria , Adolescente , Niño , Niños con Discapacidad/clasificación , Femenino , Hong Kong , Humanos , Masculino , Instituciones Académicas , Estaciones del Año
19.
IEEE Trans Neural Syst Rehabil Eng ; 27(5): 963-973, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30998471

RESUMEN

Developmental coordination disorder (DCD) is a type of motor learning difficulty that affects five to six percent of school-aged children, which may have a negative impact on the life of the sufferers. Timely and objective diagnosis of DCD are important for the success of the intervention. The present evaluation methods of DCD rely heavily on the observational analysis of occupational therapists and physiotherapists, who score the performance when children conduct some designed tasks. However, these methods are expensive, subjective, and are not easy to expand to a larger population. A fine motor evaluation system (FMES) is proposed with two views of cameras to record children's performance, when they carry out three fine motor tasks. Automated algorithms are developed to perform automated scoring of fine motor skill. The automated algorithms include task localization and individual task evaluation. The purpose of task localization is to detect each task and extract segments belonging to each task from the original video that includes multiple segments of different tasks. A convolutional neural network with temporal filtering is used to do frame-wise classification, and a boundary localization algorithm is proposed to localize each task segment. For individual task evaluation, the extracted video segments of task 1 and task 2 are evaluated based on the proposed feature extraction and time positioning algorithm, and the paper drawings of task 3 are evaluated based on image processing. The proposed methods are validated on a diverse population of children with or without DCD by comparing automated scoring with manual scoring from a professional evaluator. The experimental results suggest that the proposed methods can effectively achieve fine motor evaluation for DCD assessment. Besides, our system is a low-cost solution, and the evaluation methods developed are automated, objective, and can be suited for large population evaluation and analysis.


Asunto(s)
Trastornos de la Destreza Motora/diagnóstico , Destreza Motora , Desempeño Psicomotor/fisiología , Algoritmos , Niño , Femenino , Humanos , Masculino , Trastornos de la Destreza Motora/fisiopatología , Redes Neurales de la Computación , Reproducibilidad de los Resultados , Grabación en Video
20.
Sci Rep ; 9(1): 3015, 2019 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-30816188

RESUMEN

Tuberous Sclerosis Complex (TSC) and Lymphangioleiomyomatosis (LAM) are caused by inactivating mutations in TSC1 or TSC2, leading to mTORC1 hyperactivation. The mTORC1 inhibitors rapamycin and analogs (rapalogs) are approved for treating of TSC and LAM. Due to their cytostatic and not cytocidal action, discontinuation of treatment leads to tumor regrowth and decline in pulmonary function. Therefore, life-long rapalog treatment is proposed for the control of TSC and LAM lesions, which increases the chances for the development of acquired drug resistance. Understanding the signaling perturbations leading to rapalog resistance is critical for the development of better therapeutic strategies. We developed the first Tsc2-null rapamycin-resistant cell line, ELT3-245, which is highly tumorigenic in mice, and refractory to rapamycin treatment. In vitro ELT3-245 cells exhibit enhanced anchorage-independent cell survival, resistance to anoikis, and loss of epithelial markers. A key alteration in ELT3-245 is increased ß-catenin signaling. We propose that a subset of cells in TSC and LAM lesions have additional signaling aberrations, thus possess the potential to become resistant to rapalogs. Alternatively, when challenged with rapalogs TSC-null cells are reprogrammed to express mesenchymal-like markers. These signaling changes could be further exploited to induce clinically-relevant long-term remissions.


Asunto(s)
Resistencia a Medicamentos/genética , Mesodermo/metabolismo , Esclerosis Tuberosa/genética , Animales , Anoicis/efectos de los fármacos , Anoicis/genética , Carcinogénesis/genética , Carcinogénesis/metabolismo , Línea Celular , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Supervivencia Celular/genética , Resistencia a Medicamentos/efectos de los fármacos , Humanos , Linfangioleiomiomatosis/tratamiento farmacológico , Linfangioleiomiomatosis/genética , Linfangioleiomiomatosis/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Mesodermo/efectos de los fármacos , Ratones , Mutación/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Sirolimus/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA