Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Pharmacology ; 108(6): 540-549, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37751720

RESUMEN

INTRODUCTION: The aims of the study were to investigate the risk factors of tigecycline-induced hypofibrinogenemia and to evaluate the safety of tigecycline with concomitant antithrombotic drugs. METHODS: We performed a retrospective analysis of patients who received tigecycline for more than 3 days between January 2015 and June 2019. Clinical and laboratory data were collected including fibrinogen concertation, tigecycline dose, duration of treatment, disease severity, complete blood count, indicators of infection, liver and renal function. Risk factors of hypofibrinogenemia were analyzed by univariate and multivariate analysis. To evaluate the safety of tigecycline and concomitant antithrombotic drugs, bleeding events were assessed by comparing the decline in hemoglobin and the amount of red blood cell transfusion in patients with antithrombotic drugs and those without. RESULTS: This study included a total of 68 cases, 20 of which experienced hypofibrinogenemia while receiving tigecycline treatment. Duration of treatment, cefoperazone/sulbactam combination therapy, and fibrinogen levels prior to initiation of tigecycline were risk factors associated with tigecycline-induced hypofibrinogenemia. There were 26 recorded bleeding incidents, 25 of which happened before the start of tigecycline. Antithrombotic and non-antithrombotic patients did not differ in their hemoglobin decline or need for red blood cell transfusions while taking tigecycline. CONCLUSION: A longer treatment duration, cefoperazone/sulbactam combination therapy, and a lower level of fibrinogen before tigecycline were associated with an increased risk of tigecycline-induced hypofibrinogenemia. A combination of antithrombotic drugs and tigecycline did not aggravate the bleeding events during tigecycline treatment.


Asunto(s)
Afibrinogenemia , Antibacterianos , Humanos , Tigeciclina/efectos adversos , Antibacterianos/efectos adversos , Estudios Retrospectivos , Fibrinolíticos/efectos adversos , Cefoperazona/efectos adversos , Sulbactam/efectos adversos , Afibrinogenemia/inducido químicamente , Afibrinogenemia/tratamiento farmacológico , Hemorragia/inducido químicamente , Fibrinógeno/efectos adversos , Hemoglobinas
2.
Pharmacology ; 107(9-10): 524-536, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35820375

RESUMEN

INTRODUCTION: The purpose of this study was to assess clinical characteristics and risk factors for tigecycline-associated prothrombin time (PT) and activated partial thromboplastin time (aPTT) prolongation. METHODS: We performed a retrospective analysis on coagulation parameters before and during tigecycline treatment in 55 patients in our hospital with severe infections, mainly pneumonia caused by Acinetobacter baumannii. Patients were divided into different groups according to prolongation of PT and aPTT, and clinical features involved were explored. Univariate and multivariable binary logistic regression analyses were used to identify risk factors for tigecycline-associated PT and aPTT increase. RESULTS: We found that PT values increased from 12.73 ± 1.87 to 13.86 ± 2.06 during the treatment compared with premedication (p < 0.001), and the aPTT level prolonged significantly from 33.63 ± 11.24 to 38.15 ± 11.81 (p < 0.001). The multivariate analyses identified 2 variables that were associated with tigecycline-induced PT prolongation: albumin level (p = 0.018) and weight-adjusted tigecycline dosage (p = 0.005). In addition, treatment duration was the only risk factor for tigecycline-induced aPTT prolongation (p = 0.043). CONCLUSION: Albumin level, weight-adjusted tigecycline dosage, treatment duration may serve as risk indicators for tigecycline-associated coagulation dysfunction. Physicians should be careful with coagulation disorder when prescribing tigecycline in clinical practice, especially in patients with risk factors.


Asunto(s)
Trastornos de la Coagulación Sanguínea , Albúminas , Trastornos de la Coagulación Sanguínea/inducido químicamente , Humanos , Tiempo de Tromboplastina Parcial , Estudios Retrospectivos , Tigeciclina/efectos adversos
3.
Genes Dev ; 28(22): 2547-63, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25403183

RESUMEN

Control of Gli function by Suppressor of Fused (Sufu), a major negative regulator, is a key step in mammalian Hedgehog (Hh) signaling, but how this is achieved in the nucleus is unknown. We found that Hh signaling results in reduced Sufu protein levels and Sufu dissociation from Gli proteins in the nucleus, highlighting critical functions of Sufu in the nucleus. Through a proteomic approach, we identified several Sufu-interacting proteins, including p66ß (a member of the NuRD [nucleosome remodeling and histone deacetylase] repressor complex) and Mycbp (a Myc-binding protein). p66ß negatively and Mycbp positively regulate Hh signaling in cell-based assays and zebrafish. They function downstream from the membrane receptors, Patched and Smoothened, and the primary cilium. Sufu, p66ß, Mycbp, and Gli are also detected on the promoters of Hh targets in a dynamic manner. Our results support a new model of Hh signaling in the nucleus. Sufu recruits p66ß to block Gli-mediated Hh target gene expression. Meanwhile, Mycbp forms a complex with Gli and Sufu without Hh stimulation but remains inactive. Hh pathway activation leads to dissociation of Sufu/p66ß from Gli, enabling Mycbp to promote Gli protein activity and Hh target gene expression. These studies provide novel insight into how Sufu controls Hh signaling in the nucleus.


Asunto(s)
Regulación de la Expresión Génica , Proteínas Hedgehog/fisiología , Proteínas Represoras/metabolismo , alfa-Amilasas Salivales/metabolismo , Transducción de Señal , Animales , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Técnicas de Silenciamiento del Gen , Células HEK293 , Proteínas Hedgehog/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Mutación , Células 3T3 NIH , Unión Proteica , Proteómica , Proteínas Represoras/genética , alfa-Amilasas Salivales/genética , Pez Cebra/genética , Proteína con Dedos de Zinc GLI1
4.
World J Microbiol Biotechnol ; 37(4): 60, 2021 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-33709221

RESUMEN

The accumulation of acetate in Escherichia coli inhibits cell growth and desired protein synthesis, and cell density and protein expression are increased by reduction of acetate excretion. Dissolved oxygen (DO) is an important parameter for acetate synthesis, and the accumulation of acetate is inversely correlated to DO level. In this study, the effect of DO levels on glutamate dehydrogenase (GDH) expression was investigated, and then different DO control strategies were tested for effects on GDH expression. DO control strategy IV (50% 0-9 h, 30% 9-18 h) provided the highest cell density (15.43 g/L) and GDH concentration (3.42 g/L), values 1.59- and 1.99-times higher than those achieved at 10% DO. The accumulation of acetate was 2.24 g/L with DO control strategy IV, a decrease of 40.74% relative to that achieved for growth at 10% DO. Additionally, under DO control strategy IV, there was lower expression of PoxB, a key enzyme for acetate synthesis, at both the transcriptional and translational level. At the same time, higher transcription and protein expression levels were observed for a glyoxylate shunt gene (aceA), an acetate uptake gene (acs), gluconeogensis and anaplerotic pathways genes (pckA, ppsA, ppc, and sfcA), and a TCA cycle gene (gltA). The flux of acetate with DO strategy IV was 8.4%, a decrease of 62.33% compared with the flux at 10% DO. This decrease represents both lower flux for acetate synthesis and increased flux of reused acetate.


Asunto(s)
Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Glutamato Deshidrogenasa/genética , Glutamato Deshidrogenasa/metabolismo , Oxígeno/metabolismo , Streptococcus suis/enzimología , Streptococcus suis/metabolismo , Acetatos/metabolismo , Ciclo del Ácido Cítrico , Proteínas de Escherichia coli , Fermentación , Perfilación de la Expresión Génica , Análisis de Flujos Metabólicos , Transcriptoma
5.
Stem Cells ; 36(3): 377-391, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29148109

RESUMEN

Production of an appropriate number of distinct cell types in precise locations during embryonic development is critical for proper tissue function. Homeostatic renewal or repair of damaged tissues in adults also requires cell expansion and transdifferentiation to replenish lost cells. However, the responses of diverse cell types to tissue injury are not fully elucidated. Moreover, the molecular mechanisms underlying transdifferentiation remain poorly understood. This knowledge is essential for harnessing the regenerative potential of individual cell types. This study investigated the fate of pulmonary neuroendocrine cells (PNECs) following lung damage to understand their plasticity and potential. PNECs are proposed to carry out diverse physiological functions in the lung and can also be the cells of origin of human small cell lung cancer. We found that Notch signaling is activated in proliferating PNECs in response to epithelial injury. Forced induction of high levels of Notch signaling in PNECs in conjunction with lung injury results in extensive proliferation and transdifferentiation of PNECs toward the fate of club cells, ciliated cells and goblet cells. Conversely, inactivating Notch signaling in PNECs abolishes their ability to switch cell fate following lung insult. We also established a connection between PNEC transdifferentiation and epigenetic modification mediated by the polycomb repressive complex 2 and inflammatory responses that involve the IL6-STAT3 pathway. These studies not only reveal a major pathway that controls PNEC fate change following lung injury but also provide tools to uncover the molecular basis of cell proliferation and fate determination in response to lung injury. Stem Cells 2018;36:377-391.


Asunto(s)
Diferenciación Celular/fisiología , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Pulmón/citología , Pulmón/metabolismo , Células Neuroendocrinas/citología , Células Neuroendocrinas/metabolismo , Receptores Notch/metabolismo , Proliferación Celular/fisiología , Humanos , Transducción de Señal/fisiología
6.
J Biol Chem ; 292(9): 3888-3899, 2017 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-28119454

RESUMEN

Among the four different types of thyroid cancer, treatment of medullary thyroid carcinoma poses a major challenge because of its propensity of early metastasis. To further investigate the molecular mechanisms of medullary thyroid carcinoma and discover candidates for targeted therapies, we developed a new mouse model of medullary thyroid carcinoma based on our CGRPCreER mouse line. This system enables gene manipulation in parafollicular C cells in the thyroid, the purported cells of origin of medullary thyroid carcinoma. Selective inactivation of tumor suppressors, such as p53, Rb, and Pten, in mature parafollicular C cells via an inducible Cre recombinase from CGRPCreER led to development of murine medullary thyroid carcinoma. Loss of Pten accelerated p53/Rb-induced medullary thyroid carcinoma, indicating interactions between pathways controlled by tumor suppressors. Moreover, labeling differentiated parafollicular C cells by CGRPCreER allows us to follow their fate during malignant transformation to medullary thyroid tumor. Our findings support a model in which mutational events in differentiated parafollicular C cells result in medullary thyroid carcinoma. Through expression analysis including RNA-Seq, we uncovered major signaling pathways and networks that are perturbed following the removal of tumor suppressors. Taken together, these studies not only increase our molecular understanding of medullary thyroid carcinoma but also offer new candidates for designing targeted therapies or other treatment modalities.


Asunto(s)
Carcinoma Neuroendocrino/genética , Genes Supresores de Tumor , Neoplasias de la Tiroides/genética , Alelos , Animales , Calcitonina/metabolismo , Péptido Relacionado con Gen de Calcitonina/genética , Carcinoma Neuroendocrino/patología , Diferenciación Celular , Línea Celular , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Femenino , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Integrasas/metabolismo , Masculino , Ratones , Ratones Noqueados , Microscopía Fluorescente , Células Neuroendocrinas/metabolismo , Análisis de Secuencia de ARN , Transducción de Señal , Glándula Tiroides/metabolismo , Neoplasias de la Tiroides/patología
7.
Dev Biol ; 403(1): 101-13, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25912685

RESUMEN

Hippo signaling is a critical player in controlling the growth of several tissues and organs in diverse species. The current model of Hippo signaling postulates a cascade of kinase activity initiated by the MST1/2 kinases in response to external stimuli. This leads to inactivation of the transcriptional coactivators, YAP/TAZ, due to their cytoplasmic retention and degradation that is correlated with YAP/TAZ phosphorylation. In most tissues examined, YAP plays a more dominant role than TAZ. Whether a conserved Hippo pathway is utilized during lung growth and development is unclear. In particular, the regulatory relationship between MST1/2 and YAP/TAZ in the lung remains controversial. By employing the Shh-Cre mouse line to efficiently inactivate genes in the lung epithelium, we show that loss of MST1/2 kinases in the epithelium can lead to neonatal lethality caused by lung defects. This is manifested by perturbation of lung epithelial cell proliferation and differentiation. These phenotypes are more severe than those produced by Nkx2.1-Cre, highlighting the effects of differential Cre activity on phenotypic outcomes. Importantly, expression of YAP targets is upregulated and the ratio of phospho-YAP to total YAP protein levels is reduced in Mst1/2-deficient lungs, all of which are consistent with a negative role of MST1/2 in controlling YAP function. This model gains further support from both in vivo and in vitro studies. Genetic removal of one allele of Yap or one copy of both Yap and Taz rescues neonatal lethality and lung phenotypes due to loss of Mst1/2. Moreover, knockdown of Yap in lung epithelial cell lines restores diminished alveolar marker expression caused by Mst1/2 inactivation. These results demonstrate that MST1/2 inhibit YAP/TAZ activity and establish a conserved MST1/2-YAP axis in coordinating lung growth during development.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Pulmón/crecimiento & desarrollo , Fosfoproteínas/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Aciltransferasas , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas de Ciclo Celular , Diferenciación Celular/genética , Línea Celular , Proliferación Celular/genética , Vía de Señalización Hippo , Pulmón/embriología , Ratones , Ratones Endogámicos C57BL , Organogénesis/genética , Fosfoproteínas/metabolismo , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño , Mucosa Respiratoria/citología , Serina-Treonina Quinasa 3 , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Señalizadoras YAP
8.
Dev Biol ; 392(2): 324-33, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24886827

RESUMEN

Mammalian Hedgehog (Hh) signaling relies on three Gli transcription factors to mediate Hh responses. This process is controlled in part by a major negative regulator, Sufu, through its effects on Gli protein level, distribution and activity. In this report, we showed that Sufu regulates Gli1 protein levels by antagonizing Numb/Itch. Otherwise, Numb/Itch would induce Gli1 protein degradation. This is in contrast to inhibition of Spop-mediated degradation of Gli2/3 by Sufu. Thus, controlling protein levels of all three Gli genes by Sufu is a conserved mechanism to modulate Hh responses albeit via distinct pathways. These findings in cell-based assays were further validated in vivo. In analyzing how Sufu controls Gli proteins in different tissues, we discovered that loss of Sufu in the lung exerts different effects on Hh target genes. Hh targets Ptch1/Hhip are upregulated in Sufu-deficient lungs, consistent with Hh pathway activation. Surprisingly, protein levels of Hh target Gli1 are reduced. We also found that myofibroblasts are absent from many prospective alveoli of Sufu-deficient lungs. Myofibroblast development is dependent on PDGF signaling. Interestingly, analysis of the Pdgfra promoter revealed a canonical Gli-binding site where Gli1 resides. These studies support a model in which loss of Sufu contributes to compromised Pdgfra activation and disrupts myofibroblast development in the lung. Our work illustrates the unappreciated complexity of Hh responses where distinct Hh targets could respond differently depending on the availability of Gli proteins that control their expression.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Factores de Transcripción de Tipo Kruppel/metabolismo , Pulmón/metabolismo , Miofibroblastos/fisiología , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal/fisiología , Animales , Western Blotting , Inmunoprecipitación de Cromatina , Cartilla de ADN/genética , Regulación del Desarrollo de la Expresión Génica/genética , Células HEK293 , Proteínas Hedgehog/metabolismo , Técnicas Histológicas , Humanos , Inmunoprecipitación , Hibridación in Situ , Luciferasas , Ratones , Ratones Noqueados , Microscopía Fluorescente , Modelos Biológicos , Reacción en Cadena de la Polimerasa , ARN Interferente Pequeño/genética , Proteínas Represoras/genética , Proteína con Dedos de Zinc GLI1
9.
Dig Dis Sci ; 60(12): 3513-24, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26112990

RESUMEN

Hepatitis B virus (HBV) infection is a major etiology of chronic liver disease worldwide. In the past decade, nonalcoholic fatty liver disease (NAFLD) has emerged as a common liver disorder in general population. Accordingly, the patient number of chronic hepatitis B (CHB) concomitant with NAFLD grows rapidly. The present article reviewed the recent studies aiming to explore the relationship between CHB and NAFLD from different aspects, including the relevant pathogenesis of CHB and NAFLD, the intracellular molecular mechanisms overlaying HBV infection and hepatic steatosis, and the observational studies with animal models and clinical cohorts for analyzing the coincidence of the two diseases. It is concluded that although numerous cross-links have been suggested between the molecular pathways in HBV infection and NAFLD pathogenesis, regarding whether HBV infection can substantially interfere with the occurrence of NAFLD or vice versa in the patients, there is still far from a conclusive agreement.


Asunto(s)
Hepatitis B Crónica/complicaciones , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Animales , Virus de la Hepatitis B , Hepatitis B Crónica/virología , Hepatitis Viral Animal/complicaciones , Humanos , Enfermedad del Hígado Graso no Alcohólico/etiología
10.
Proc Natl Acad Sci U S A ; 109(43): 17531-6, 2012 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-23047698

RESUMEN

Pulmonary neuroendocrine cells (PNECs) are proposed to be the first specialized cell type to appear in the lung, but their ontogeny remains obscure. Although studies of PNECs have suggested their involvement in a number of lung functions, neither their in vivo significance nor the molecular mechanisms underlying them have been elucidated. Importantly, PNECs have long been speculated to constitute the cells of origin of human small-cell lung cancer (SCLC) and recent mouse models support this hypothesis. However, a genetic system that permits tracing the early events of PNEC transformation has not been available. To address these key issues, we developed a genetic tool in mice by introducing a fusion protein of Cre recombinase and estrogen receptor (CreER) into the calcitonin gene-related peptide (CGRP) locus that encodes a major peptide in PNECs. The CGRP(CreER) mouse line has enabled us to manipulate gene activity in PNECs. Lineage tracing using this tool revealed the plasticity of PNECs. PNECs can be colabeled with alveolar cells during lung development, and following lung injury, PNECs can contribute to Clara cells and ciliated cells. Contrary to the current model, we observed that elimination of PNECs has no apparent consequence on Clara cell recovery. We also created mouse models of SCLC in which CGRP(CreER) was used to ablate multiple tumor suppressors in PNECs that were simultaneously labeled for following their fate. Our findings suggest that SCLC can originate from differentiated PNECs. Together, these studies provide unique insight into PNEC lineage and function and establish the foundation of investigating how PNECs contribute to lung homeostasis, injury/repair, and tumorigenesis.


Asunto(s)
Neoplasias Pulmonares/patología , Pulmón/citología , Células Neuroendocrinas/fisiología , Animales , Transformación Celular Neoplásica , Genes de Retinoblastoma , Genes p53 , Humanos , Ratones , Fosfohidrolasa PTEN/genética
11.
Dev Dyn ; 242(11): 1307-19, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23907739

RESUMEN

BACKGROUND: Motile cilia on the inner lining of the oviductal epithelium play a central role in ovum transport toward the uterus and subsequent fertilization by sperm. While the basic ultrastructure of 9+2 motile cilia (nine peripheral microtubule doublets surrounding a central pair) has been characterized, many important steps of ciliogenesis remain poorly understood. RESULTS: Our previous studies on mammalian Fused (Fu) (Stk36), a putative serine-threonine kinase, reveal a critical function of Fu in central pair construction and cilia orientation of motile cilia that line the tracheal and ependymal epithelia. These findings identify a novel regulatory component for these processes. In this study, we show that Fu is expressed in the multi-ciliated oviductal epithelium in several vertebrates, suggesting a conserved function of Fu in the oviduct. In support of this, analysis of Fu-deficient mouse oviducts uncovers a similar role of Fu in central pair construction and cilia orientation. We also demonstrate that Fu localizes to motile cilia and physically associates with kinesin Kif27 located at the cilium base and known central pair components Spag16 and Pcdp1. CONCLUSIONS: Our results delineate a novel pathway for central pair apparatus assembly and add important insight to the biogenesis and function of oviductal motile cilia.


Asunto(s)
Cilios/metabolismo , Oviductos/embriología , Oviductos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Línea Celular , Cilios/ultraestructura , Femenino , Humanos , Hibridación in Situ , Mamíferos/embriología , Mamíferos/metabolismo , Ratones , Ratones Mutantes , Microscopía Electrónica de Transmisión , Proteínas Serina-Treonina Quinasas/genética
12.
J Cancer ; 15(8): 2329-2339, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38495491

RESUMEN

Lung squamous cell carcinoma (LUSC) accounts for approximately 25% to 30% of lung cancers, but largely no targeted therapy is available against it, calling for identification of new oncogenes in LUSC growth for new therapeutic targets. In this study, REL was identified through a screening for oncogenes that are highly amplified in human LUSC. Its expression was associated with poor prognosis in LUSC patients. Furthermore, knockdown of c-Rel in LUSC cell lines lead to significant decrease in cell proliferation and migration. Mechanistically, c-Rel knockdown suppressed NFκB pathway by blocking phosphorylation of IκB. Consistently, pharmaceutic inhibition of c-Rel also. In orthotopic xenograft lung cancer mouse model, c-Rel knockdown inhibited the tumor growth. Cancer cell proliferation and epithelial-mesenchymal-transition (EMT) of the tumors were impaired by c-Rel knockdown. Finally, it's confirmed in precision-cut tumor slices of LUSC that deletion of c-Rel inhibits the NFκB pathway and cancer cell growth. Accordingly, we hypothesize that c-Rel promotes the activation of the NFκB pathway by promoting the phosphorylation of IκB in LUSC. Our study reveals REL as a novel LUSC oncogene and provides new insights into the molecular regulation of LUSC, which will provide new therapeutic targets for the treatment of squamous lung cancer.

13.
Stem Cells Dev ; 2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39119800

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a progressive lung disease with no cure except transplantation. Abnormal alveolar epithelial regeneration is a key driver of IPF development. The function of Yes1 Associated Transcriptional Regulator (YAP) in alveolar regeneration and IPF pathogenesis remains elusive. Here, we first revealed the activation of YAP in alveolar epithelium 2 cells (AEC2s) from human IPF lungs and fibrotic mouse lungs. Notably, conditional deletion of YAP in mouse AEC2s exacerbated bleomycin-induced pulmonary fibrosis. Intriguingly, we showed in both conditional knockout mice and alveolar organoids that YAP deficiency impaired AEC2 proliferation and differentiation into alveolar epithelium 1 cells (AEC1s). Mechanistically, YAP regulated expression levels of genes associated with cell cycle progression and AEC1 differentiation. Furthermore, overexpression of YAP in vitro promoted cell proliferation. These results indicate the critical role of YAP in alveolar regeneration and IPF pathogenesis. Our findings provide new insights into the regulation of alveolar regeneration and IPF pathogenesis, paving the road for developing novel treatment strategies.

14.
Int J Biol Macromol ; 253(Pt 5): 127202, 2023 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-37793530

RESUMEN

The typical function of Drosha is participating in cleaving pri-miRNA, the initial step of miRNA biogenesis, in the nucleus. Since Drosha has a double-stranded RNA-binding domain and two RNase III domains, when it binds and/or cleaves other RNA species other than pri-miRNA, Drosha is able to induce a variety of novel biological effects. Moreover, by interacting with other protein, Drosha is able to modify the function of other protein complexes. Recently, diverse non-classical functions of Drosha have been demonstrated, such as promoting DNA damage repair, transcriptional activation and inhibition, pre-mRNA splicing regulation, mRNA destabilization, and virus-host interaction. In this review, we describe these newly discovered functions of Drosha in order to present a panoramic picture of the novel biological processes that Drosha is involved in.


Asunto(s)
MicroARNs , Ribonucleasa III , Ribonucleasa III/genética , Ribonucleasa III/metabolismo , MicroARNs/genética , Proteínas/metabolismo , Regulación de la Expresión Génica , Procesamiento Postranscripcional del ARN
15.
Sci Data ; 10(1): 35, 2023 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-36653358

RESUMEN

Data of the diabetes mellitus patients is essential in the study of diabetes management, especially when employing the data-driven machine learning methods into the management. To promote and facilitate the research in diabetes management, we have developed the ShanghaiT1DM and ShanghaiT2DM Datasets and made them publicly available for research purposes. This paper describes the datasets, which was acquired on Type 1 (n = 12) and Type 2 (n = 100) diabetic patients in Shanghai, China. The acquisition has been made in real-life conditions. The datasets contain the clinical characteristics, laboratory measurements and medications of the patients. Moreover, the continuous glucose monitoring readings with 3 to 14 days as a period together with the daily dietary information are also provided. The datasets can contribute to the development of data-driven algorithms/models and diabetes monitoring/managing technologies.


Asunto(s)
Automonitorización de la Glucosa Sanguínea , Diabetes Mellitus , Humanos , Algoritmos , Glucemia , China , Aprendizaje Automático
16.
Life Sci ; 321: 121578, 2023 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-36958438

RESUMEN

AIMS: Lung squamous cell carcinoma (LUSC) causes over 400,000 deaths annually, yet it lacks targeted therapy. A major antagonist of Hedgehog pathway, HHIP (Hedgehog Interacting Protein) plays an important role in LUSC; however, the regulatory mechanism remains unclear. Long non-coding RNA HHIP-AS1 plays suppressive or promotive roles in different cancers, but its role in LUSC remains unknown. This manuscript is to investigate regulatory mechanism of HHIP and the role of HHIP-AS1 in LUSC. MAIN METHODS: Precision-cut lung slices (PCLS) from human LUSC samples are cultured to mimic LUSC growth. Overexpression and knockdown in multiple LUSC cell lines and PCLS are achieved by lentivirus infection. Transcriptome profile and lung cancer activity are evaluated by RNA-sequencing, immunostaining and CCK8 assay etc. KEY FINDINGS: HHIP is regulated independently of Hh pathway in LUSC. Additionally, downregulation of HHIP-AS1 is associated with poor prognosis. Consistently, HHIP-AS1 inhibits LUSC growth by suppressing cell proliferation and migration. Transcriptome profiling of HHIP-AS1 knockdown (KD) cells uncovered HHIP downregulation. Interestingly, a comparison between the transcriptomes of HHIP-AS1 KD or HHIP KD cells manifested high similarity. Subsequently it's confirmed that HHIP-AS1 regulates HHIP in LUSC cells. Notably, HHIP-AS1 regulation on LUSC growth is achieved through stabilizing HHIP mRNA rather than regulating MIR-153-3P/PCDHGA9 or MIR-425-5P/DNYC1I2. Finally, it's confirmed in PCLS from human LUSC samples that HHIP-AS1 suppresses LUSC via regulating HHIP mRNA. SIGNIFICANCE: This study uncovers HHIP-AS1 as a novel tumor suppressor in LUSC and provides new insights into the molecular regulation of LUSC, which will help developing new therapeutic strategies.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Carcinoma de Células Escamosas , Neoplasias Pulmonares , MicroARNs , ARN Largo no Codificante , Humanos , MicroARNs/genética , ARN Largo no Codificante/genética , ARN Mensajero/genética , Proteínas Hedgehog/genética , Línea Celular Tumoral , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Proliferación Celular/genética , Pulmón/patología , Regulación Neoplásica de la Expresión Génica , Movimiento Celular/genética , Proteínas Portadoras/genética , Glicoproteínas de Membrana/genética
17.
Sci Transl Med ; 14(661): eabm7621, 2022 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-35579533

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus driving the ongoing coronavirus disease 2019 (COVID-19) pandemic, continues to rapidly evolve. Because of the limited efficacy of vaccination in prevention of SARS-CoV-2 transmission and continuous emergence of variants of concern (VOCs), orally bioavailable and broadly efficacious antiviral drugs are urgently needed. Previously, we showed that the parent nucleoside of remdesivir, GS-441524, has potent anti-SARS-CoV-2 activity. Here, we report that esterification of the 5'-hydroxyl moieties of GS-441524 markedly improved antiviral potency. This 5'-hydroxyl-isobutyryl prodrug, ATV006, demonstrated excellent oral bioavailability in rats and cynomolgus monkeys and exhibited potent antiviral efficacy against different SARS-CoV-2 VOCs in vitro and in three mouse models. Oral administration of ATV006 reduced viral loads and alleviated lung damage when administered prophylactically and therapeutically to K18-hACE2 mice challenged with the Delta variant of SARS-CoV-2. These data indicate that ATV006 represents a promising oral antiviral drug candidate for SARS-CoV-2.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Profármacos , Adenosina/uso terapéutico , Adenosina Monofosfato/análogos & derivados , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , Ratones , Profármacos/farmacología , Profármacos/uso terapéutico , Ratas , SARS-CoV-2
18.
Aging (Albany NY) ; 13(12): 15770-15784, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-34168096

RESUMEN

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19), and is highly contagious and pathogenic. TMPRSS2 and Neuropilin-1, the key components that facilitate SARS-CoV-2 infection, are potential targets for treatment of COVID-19. Here we performed a comprehensive analysis on NRP1 and TMPRSS2 in lung to provide information for treating comorbidity of COVID-19 with lung cancer. NRP1 is widely expressed across all the human tissues while TMPRSS2 is expressed in a restricted pattern. High level of NRP1 associates with worse prognosis in multiple cancers, while high level of TMPRSS2 is associated with better survival of Lung Adenocarcinoma (LUAD). Moreover, NRP1 positively correlates with the oncogenic Cancer Associated Fibroblast (CAF), macrophage and endothelial cells infiltration, negatively correlates with infiltration of CD8+ T cell, the tumor killer cell in Lung Squamous cell carcinoma (LUSC). TMPRSS2 shows negative correlation with the oncogenic events in LUAD. RNA-seq data show that NRP1 level is slightly decreased in peripheral blood of ICU admitted COVID-19 patients, unaltered in lung, while TMPRSS2 level is significantly decreased in lung of COVID-19 patients. Our analysis suggests NRP1 as a potential therapeutic target, while sets an alert on targeting TMPRSS2 for treating comorbidity of COVID-19 and lung cancers.


Asunto(s)
Adenocarcinoma del Pulmón/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/metabolismo , Neuropilina-1/fisiología , Serina Endopeptidasas/fisiología , Adenocarcinoma del Pulmón/mortalidad , Linfocitos T CD8-positivos/metabolismo , COVID-19/genética , COVID-19/metabolismo , Fibroblastos Asociados al Cáncer/metabolismo , Simulación por Computador , Células Endoteliales/metabolismo , Humanos , Neoplasias Pulmonares/mortalidad , Macrófagos/metabolismo , Neuropilina-1/genética , RNA-Seq , SARS-CoV-2 , Serina Endopeptidasas/genética
19.
J Diabetes Res ; 2021: 9943344, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34917687

RESUMEN

Insulin treatment was confirmed to reduce insulin resistance, but the underlying mechanism remains unknown. Caveolin-1 (Cav-1) is a functional protein of the membrane lipid rafts, known as caveolae, and is widely expressed in mammalian adipose tissue. There is increasing evidence that show the involvement of Cav-1 in the AKT activation, which is responsible for insulin sensitivity. Our aim was to investigate the effect of Cav-1 depletion on insulin sensitivity and AKT activation in glargine-treated type 2 diabetic mice. Mice were exposed to a high-fat diet and subject to intraperitoneal injection of streptozotocin to induce diabetes. Next, glargine was administered to treat T2DM mice for 3 weeks (insulin group). The expression of Cav-1 was then silenced by injecting lentiviral-vectored short hairpin RNA (shRNA) through the tail vein of glargine-treated T2DM mice (CAV1-shRNA group), while scramble virus injection was used as a negative control (Ctrl-shRNA group). The results showed that glargine was able to upregulate the expression of PI3K and activate serine phosphorylation of AKT through the upregulation of Cav-1 expression in paraepididymal adipose tissue of the insulin group. However, glargine treatment could not activate AKT pathway in Cav-1 silenced diabetic mice. These results suggest that Cav-1 is essential for the activation of AKT and improving insulin sensitivity in type 2 diabetic mice during glargine treatment.


Asunto(s)
Caveolina 1/metabolismo , Insulina Glargina/farmacología , Resistencia a la Insulina/genética , Animales , Modelos Animales de Enfermedad , Insulina Glargina/metabolismo , Resistencia a la Insulina/fisiología , Ratones , Ratones Endogámicos NOD
20.
Artículo en Inglés | MEDLINE | ID: mdl-34770016

RESUMEN

Breastfeeding is recommended over formula feeding, but human breast milk (HBM) composition varies and can be affected by food additives. Whether flame-retardant polybrominated diphenyl ethers (PBDEs) found in HBM interact with lipid components of HBM to impede infant neurodevelopment is a critical public health issue. Using lipidomic analysis, we examined the association of PBDEs in HBM and HBM lipid components with infant neurodevelopment. HBM samples (n = 100) were collected at the beginning stage of breastfeeding and analyzed for 30 PBDE congeners as well as a group of lipid components by using high-resolution gas chromatography, mass spectrometry, and liquid chromatography time-of-flight mass spectrometry. Infants were examined at 8 to 12 months of age by using the Bayley-III to assess neurodevelopment. A total of seven PBDEs, 35 lipids, and 27 fatty acids in HBM showed significant associations with Bayley-III scores. Multivariate analysis confirmed that these candidate PBDEs and lipid components were significant predictors of infant neurodevelopment. Eicosapentaenoic acid and docosapentaenoic acid in HBM showed no association with infant neurodevelopment in the general Taiwanese population. While certain PBDEs may play a role, our findings indicate that the lipid components of HBM are directly important for infant neurodevelopment.


Asunto(s)
Contaminantes Ambientales , Éteres Difenilos Halogenados , Contaminantes Ambientales/análisis , Femenino , Cromatografía de Gases y Espectrometría de Masas , Éteres Difenilos Halogenados/análisis , Humanos , Lactante , Lípidos , Leche Humana/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA