Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mol Carcinog ; 63(6): 1174-1187, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38501385

RESUMEN

Sorbin and SH3 domain-containing 2 (SORBS2) is an RNA-binding protein and has been implicated in the development of some cancers. However, its role in bladder cancer (BC) is yet to be established. The expression of SORBS2 in BC tissues was determined from the Gene Expression Omnibus and Gene Expression Profiling Interactive Analysis databases and collected paired tumor/normal samples. The effects of SORBS2 on BC cells were detected by CCK-8, colony formation, Transwell, dual-luciferase, RNA immunoprecipitation, chromatin immunoprecipitation, and DNA pull-down assays. In vivo, BC cell growth and metastasis were studied by a xenograft subcutaneous model and a tail-vein metastasis model. The results showed that SORBS2 expression was significantly decreased in BC tissues and cells. SORBS2 overexpression inhibited cell proliferation, migration, invasion, and epithelial-mesenchymal transition in vitro and tumor growth and metastasis in vivo, while silencing SORBS2 produced the opposite effect. Mechanistically, we found that SORBS2 enhanced the stability of tissue factor pathway inhibitor (TFPI) mRNA via direct binding to its 3' UTR. Restoration of TFPI expression reversed SORBS2 knockdown-induced malignant phenotypes of BC cells. In addition, SORBS2 expression was negatively regulated by the transcription factor specificity protein 1 (SP1). Conversely, SORBS2 can be transcriptionally regulated by SP1 and inhibit BC cell growth and metastasis via stabilization of TFPI mRNA, indicating SORBS2 may be a promising therapeutic target for BC.


Asunto(s)
Movimiento Celular , Proliferación Celular , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Proteínas de Unión al ARN , Neoplasias de la Vejiga Urinaria , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/patología , Neoplasias de la Vejiga Urinaria/metabolismo , Humanos , Animales , Ratones , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/genética , Ratones Desnudos , Femenino , Masculino , Ensayos Antitumor por Modelo de Xenoinjerto , Ratones Endogámicos BALB C
2.
Sci Rep ; 13(1): 18882, 2023 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-37919345

RESUMEN

The Myodural Bridge (MDB) is a physiological structure that is highly conserved in mammals and many of other tetrapods. It connects the suboccipital muscles to the cervical spinal dura mater (SDM) and transmits the tensile forces generated by the suboccipital muscles to the SDM. Consequently, the MDB has broader physiological potentials than just fixing the SDM. It has been proposed that MDB significantly contributes to the dynamics of cerebrospinal fluid (CSF) movements. Animal models of suboccipital muscle atrophy and hyperplasia were established utilizing local injection of BTX-A and ACE-031. In contrast, animal models with surgical severance of suboccipital muscles, and without any surgical operation were set as two types of negative control groups. CSF secretion and reabsorption rates were then measured for subsequent analysis. Our findings demonstrated a significant increase in CSF secretion rate in rats with the hyperplasia model, while there was a significant decrease in rats with the atrophy and severance groups. We observed an increase in CSF reabsorption rate in both the atrophy and hyperplasia groups, but no significant change was observed in the severance group. Additionally, our immunohistochemistry results revealed no significant change in the protein level of six selected choroid plexus-CSF-related proteins among all these groups. Therefore, it was indicated that alteration of MDB-transmitted tensile force resulted in changes of CSF secretion and reabsorption rates, suggesting the potential role that MDB may play during CSF circulation. This provides a unique research insight into CSF dynamics.


Asunto(s)
Duramadre , Músculos del Cuello , Animales , Ratas , Hiperplasia , Duramadre/fisiología , Músculos del Cuello/fisiología , Movimiento , Mamíferos , Atrofia , Líquido Cefalorraquídeo
3.
Cancer Lett ; 556: 216058, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36627049

RESUMEN

One of the most abundant protein-protein interaction domains in the human proteome is the WD40 repeat (WDR) domain. A Gene Expression Omnibus dataset revealed 37 differentially expressed WDR domain genes in bladder cancer (BC). WD repeat domain 54 (WDR54), an upregulated WDR domain gene, was selected for further investigation. Sixty pairs of frozen BC tumor and non-malignant bladder tissues and 83 paraffin-embedded BC tissue specimens were obtained. Loss-/gain-of-function experiments were carried out using BC and xenograft tumor models. WDR54 was overexpressed in BC cells, and its high expression was linked to tumor stage and lymph node metastases in patients. WDR54 contributed to the tumorigenesis and metastasis of BC and impaired its chemosensitivity. WDR54 prevented the degradation and ubiquitination of the mediator of ErbB2-driven cell motility 1 (MEMO1). WDR54 also promoted the interaction between MEMO1 and insulin receptor substrate 1 (IRS1) and activated the IRS1/AKT/ß-catenin pathway in BC cells. Particularly, WDR54 depended on MEMO1 to exert its biological functions. Our study demonstrated the relevance of WDR54 in BC and provides insight into the molecular mechanism underlying BC.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular , Neoplasias de la Vejiga Urinaria , Humanos , Carcinogénesis/genética , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Transformación Celular Neoplásica/genética , Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/metabolismo , Repeticiones WD40
4.
PLoS One ; 17(9): e0273193, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36054096

RESUMEN

During mammalian evolution, the Myodural Bridges (MDB) have been shown to be highly conserved anatomical structures. However, the putative physiological function of these structures remains unclear. The MDB functionally connects the suboccipital musculature to the cervical spinal dura mater, while passing through the posterior atlanto-occipital and atlanto-axial interspaces. MDB transmits the tensile forces generated by the suboccipital muscles to the cervical dura mater. Moreover, head movements have been shown to be an important contributor to human CSF circulation. In the present study, a 16-week administration of a Myostatin-specific inhibitor, ACE-031, was injected into the suboccipital musculature of rats to establish an experimental animal model of hyperplasia of the suboccipital musculature. Using an optic fiber pressure measurement instrument, the present authors observed a significant increase in intracranial pressure (ICP) while utilizing the hyperplasia model. In contrast, surgically severing the MDB connections resulted in a significant decrease in intracranial pressure. Thus, these results indicated that muscular activation of the MDB may affect CSF circulation, suggesting a potential functional role of the MDB, and providing a new research perspective on CSF dynamics.


Asunto(s)
Presión Intracraneal , Músculos del Cuello , Animales , Duramadre/fisiología , Humanos , Hiperplasia , Mamíferos , Cuello , Ratas
5.
Urol Oncol ; 40(8): 382.e15-382.e24, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35637063

RESUMEN

PURPOSE: Bladder cancer is a kind of common malignant cancer in the urinary system. The expression of EDARADD (ectodysplasin-A receptor-associated death domain) in bladder cancer is higher than the normal samples. However, its role in bladder cancer remains unknown. In the present study, we analyzed the expression of EDARADD in 81 bladder cancer samples by immunohistochemistry as well as its correlation with clinical characteristics. In addition, the role of EDARADD was also explored through loss of function. MATERIALS AND METHODS: Cell proliferation assay and MTT assay were conducted to assess the proliferation of bladder cancer cells and transwell assay and wound healing assay were conducted to assess the migration of bladder cancer cells. On the other hand, the levels of epithelial-mesenchymal transition (EMT) associated proteins and the key molecules in the MAPK signaling pathway were detected by western blot. In vivo experiments were also conducted to determine the effect of EDARADD silencing on the metastasis of bladder cancer cells and the MAPK signaling pathway. RESULTS: EDARADD was highly expressed in bladder cancer samples, especially in high-grade bladder cancer samples. The high EDARADD level indicated a poor survival. Interestingly, EDARADD silencing suppressed the proliferation, migration and EMT of bladder cancer cells. Furthermore, the MAPK signaling pathway was repressed by EDARADD silencing. Additionally, silencing EDARADD also inhibited the metastasis of bladder cancer and the MAPK signaling pathway in vivo. It is indicated that silencing EDARADD may suppress the proliferation and metastasis of bladder cancer cells through the MAPK signaling pathway. CONCLUSION: These results indicate that EDARADD may become a probable target for the treatment of bladder cancer.


Asunto(s)
Neoplasias de la Vejiga Urinaria , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Proteína de Dominio de Muerte Asociada a Edar/metabolismo , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Invasividad Neoplásica/genética , Neoplasias de la Vejiga Urinaria/patología
6.
Bioact Mater ; 13: 269-285, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35224308

RESUMEN

The efficacious delivery of antimicrobial drugs to intractable oral biofilms remains a challenge due to inadequate biofilm penetration and lack of pathogen targeting. Herein, we have developed a microenvironment-activated poly(ethylene glycol) (PEG)-sheddable nanoplatform to mediate targeted delivery of drugs into oral biofilms for the efficient prevention of dental caries. The PEGylated nanoplatform with enhanced biofilm penetration is capable of deshielding the PEG layer under slightly acidic conditions in a PEG chain length-dependent manner to re-expose the bacteria-targeting ligands, thereby facilitating targeted codelivery of ciprofloxacin (CIP) and IR780 to the bacteria after accumulation within biofilms. The nanoplatform tends to induce bacterial agglomeration and suffers from degradation in the acidic oral biofilm microenvironment, triggering rapid drug release on demand around bacterial cells. The self-modulating nanoplatform under near-infrared (NIR) irradiation accordingly displays greatly augmented potency in oral biofilm penetration and disruption compared with drugs alone. Topical oral treatment with nanoplatforms involving synergetic pharmacological and photothermal/photodynamic trinary therapy results in robust biofilm dispersion and efficacious suppression of severe tooth decay in rats. This versatile nanoplatform can promote local accumulation and specific drug transport into biofilms and represents a new paradigm for targeted drug delivery for the management of oral biofilm-associated infections.

7.
DNA Cell Biol ; 41(2): 179-189, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35007433

RESUMEN

Bladder cancer (BC) is the most common type of malignant tumor in the genitourinary system. Through the microarray analysis of clinical samples, long noncoding RNA HAND2-AS1 expression was found to be downregulated in BC tissues. However, the function of HAND2-AS1 on BC and underlying mechanism are unclear. In this study, the correlations of HAND2-AS1 with clinicopathological parameters in BC patients were determined. The gain- and loss-of-function experiments were conducted to examine the role of HAND2-AS1 in malignant behaviors of BC cells in vitro and in vivo. Then, we paid attention to miR-17-5p/KLF9 axis to illustrate the molecular mechanism. Results showed that HAND2-AS1 was downregulated in BC tissues, and its overexpression significantly inhibited cell proliferation, migration, and invasion in vitro, as well as tumor growth in vivo. Knockdown of HAND2-AS1 caused an opposite effect on BC cell malignancies. Furthermore, miR-17-5p was shown to be a direct target of HAND2-AS1, and it reversed the inhibitory effect of HAND2-AS1 on BC malignancies. Also, as a downstream factor of miR-17-5p, KLF9 silencing was demonstrated to mediate the role of miR-17-5p inhibitor in BC cell proliferation and invasion. Thus, it suggests that HAND2-AS1 acts as a suppressor in BC development through miR-17-5p/KLF9 axis.


Asunto(s)
Neoplasias de la Vejiga Urinaria
8.
Biomaterials ; 280: 121312, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34896861

RESUMEN

Checkpoint inhibitors, such as antibodies blocking the PD-1/PD-L1 pathway, are among the most promising immunotherapies to treat metastatic cancers, but their response rate remains low. In addition, the usage of monoclonal antibodies as checkpoint inhibitors is associated with a series of drawbacks. Herein, an all synthetic nanoparticle with PD-L1 blockade capability is developed for cancer photothermal-immunotherapy. The polymeric nanoparticle integrates photothermal treatment, antitumor vaccination, and PD-1/PD-L1 blockade in a single system to augment the antitumor efficacy. In a CT26 bilateral tumor model, intravenously injected nanoparticles accumulate in tumor sites and mediate strong photothermal effects, eradicate the NIR treated primary tumors and elicit strong antitumor immunity by inducing immunogenic cell death (ICD). Growth of the untreated distant tumors is also suppressed due to the synergies of systemic antitumor immune activation and PD-L1 blockade. Our strategy offers a simple but promising approach for the treatment of metastatic cancer.


Asunto(s)
Nanopartículas , Neoplasias , Anticuerpos Monoclonales , Antígeno B7-H1 , Línea Celular Tumoral , Humanos , Inmunoterapia , Neoplasias/terapia
9.
Biomaterials ; 280: 121309, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34896862

RESUMEN

Intracellular bacteria-mediated antibiotic tolerance, which acts as a "Trojan horse," plays a critical and underappreciated role in chronic and recurrent infections. Failure of conventional antibiotic therapy is often encountered because infected cells prevent drug permeation or the drug concentration is too low at the site of resident bacteria. New paradigms are therefore urgently needed for intracellular anti-infective therapy. Here, a novel therapeutic was developed for targeted delivery of antibiotics into bacteria-infected macrophages to improve drug accumulation in intracellular niches and bactericidal activity of antibiotics against intracellular pathogens. This hierarchical nanoplatform includes a glycocalyx-mimicking shell that enables rapid uptake by macrophages. Subsequently, the targeting moieties are activated in response to the bacteria, and the release of entrapped antibiotics is triggered by bacteria and bacteria-secreted enzymes. The self-immolative drug delivery nanoplatform eliminates intracellular pathogenic bacteria residing in macrophages more efficiently compared to drugs alone. The in vivo dynamically monitored nanosystem also efficiently inhibited the growth of intracellular Staphylococcus aureus in infected muscles of mice with negligible systemic toxicity. The novel dual-targeting design of an all-in-one therapeutic platform can be used as an alternative strategy to reanimate antibiotic therapy against multifarious intracellular bacterial infections.


Asunto(s)
Enfermedades Parasitarias , Infecciones Estafilocócicas , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Sistemas de Liberación de Medicamentos , Ratones , Enfermedades Parasitarias/tratamiento farmacológico , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus
10.
ACS Appl Mater Interfaces ; 13(33): 38979-38989, 2021 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-34433249

RESUMEN

Chronic infections caused by Pseudomonas aeruginosa pose severe threats to human health. Traditional antibiotic therapy has lost its total supremacy in this battle. Here, nanoplatforms activated by the clinical microenvironment are developed to treat P. aeruginosa infection on the basis of dynamic borate ester bonds. In this design, the nanoplatforms expose targeted groups for bacterial capture after activation by an acidic infection microenvironment, resulting in directional transport delivery of the payload to bacteria. Subsequently, the production of hyperpyrexia and reactive oxygen species enhances antibacterial efficacy without systemic toxicity. Such a formulation with a diameter less than 200 nm can eliminate biofilm up to 75%, downregulate the level of cytokines, and finally promote lung repair. Collectively, the biomimetic design with phototherapy killing capability has the potential to be an alternative strategy against chronic infections caused by P. aeruginosa.


Asunto(s)
Antibacterianos/química , Verde de Indocianina/química , Nanocápsulas/química , Fármacos Fotosensibilizantes/química , Polímeros/química , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones por Pseudomonas/radioterapia , Células A549 , Animales , Antibacterianos/farmacología , Biopelículas/efectos de los fármacos , Composición de Medicamentos , Liberación de Fármacos , Sinergismo Farmacológico , Humanos , Verde de Indocianina/farmacología , Rayos Infrarrojos , Masculino , Metacrilatos/química , Ratones Endogámicos BALB C , Fotoquimioterapia , Fármacos Fotosensibilizantes/farmacología , Polietilenglicoles/química , Pseudomonas aeruginosa/efectos de los fármacos
11.
ACS Appl Mater Interfaces ; 13(29): 33790-33801, 2021 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-34254513

RESUMEN

Hypoxia, a common characteristic of bacterial infections, is known to be closely associated with the emergence of multidrug-resistant bacteria, which hastens the need to develop advanced microbicides and antibacterial techniques. Photodynamic therapy is a promising strategy to reduce bacterial antibiotic resistance and employs photosensitizers, excitation light sources, and sufficient oxygen to generate toxic reactive oxygen species (ROS). The inherent limitation of PDT is that the generation of ROS is restricted by the hypoxic microenvironment in infection sites. Here, an oxygen self-supplying nanotherapeutic is developed to enhance antibacterial activity against multidrug-resistant bacteria on the basis of fluorinated boron dipyrromethene (BODIPY)-based glycomimetics. The nanotherapeutic not only could capture the bacteria efficiently but also was able to act as an oxygen carrier to relieve the hypoxic microenvironment of bacterial infections, thus achieving enhanced PDT efficacy. In a Pseudomonas aeruginosa infection of a rat cornea, typical administration of the nanotherapeutic decreased the infiltrate and showed a faster healing capacity in comparison with BODIPY-based glycomimetics. Self-supplying oxygen nanotherapeutics that relieve the hypoxic microenvironment and interfere with bacterial colonization have been shown to be a promising candidate for the management of drug-resistant microbial keratitis.


Asunto(s)
Antibacterianos/uso terapéutico , Infecciones Bacterianas del Ojo/tratamiento farmacológico , Hipoxia/tratamiento farmacológico , Queratitis/tratamiento farmacológico , Nanopartículas/uso terapéutico , Oxígeno/metabolismo , Animales , Antibacterianos/química , Antibacterianos/efectos de la radiación , Biopelículas/efectos de los fármacos , Compuestos de Boro/química , Compuestos de Boro/efectos de la radiación , Compuestos de Boro/uso terapéutico , Córnea/metabolismo , Córnea/microbiología , Córnea/patología , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Infecciones Bacterianas del Ojo/metabolismo , Infecciones Bacterianas del Ojo/patología , Hipoxia/metabolismo , Hipoxia/patología , Queratitis/metabolismo , Queratitis/patología , Luz , Ratones , Células 3T3 NIH , Nanopartículas/química , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/efectos de la radiación , Fármacos Fotosensibilizantes/uso terapéutico , Ácidos Polimetacrílicos/química , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones por Pseudomonas/metabolismo , Infecciones por Pseudomonas/patología , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/fisiología , Ratas
12.
J Mater Chem B ; 9(17): 3689-3695, 2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-33861292

RESUMEN

Exogenous reactive oxygen species (ROS) generation is a promising antibacterial strategy. The short diffusion distance coupled with the transient existence of ROS restrict their precise release at inflammation sites, so it is imperative to regulate the reactive sites of ROS donors. In this work, we developed a glycomimetic-decorated fluorescent nanobiocide to mediate the release of ROS generated from CuInS/ZnS quantum dots. The introduction of glycomimetics innovatively improved the biocompatibility of the hydrophobic quantum dots, allowing pathogenic bacteria to be targeted. The functionalized CuInS/ZnS quantum dots allowed simultaneous fluorescent reporting and sterilization under 660 nm illumination. Moreover, the nanobiocide can serve as a cell-binding glue causing bacterial aggregation, disrupting bacterial adhesion to host cells and inhibiting biofilm formation. Collectively, this work indicated the far-reaching future of ROS-generating biomimetic design for multifunctional nanobiocides to combat bacterial infections.


Asunto(s)
Antibacterianos/química , Colorantes Fluorescentes/química , Infecciones/tratamiento farmacológico , Puntos Cuánticos/química , Células 3T3 , Adhesivos/química , Animales , Antibacterianos/farmacología , Biopelículas , Cobre/química , Eritrocitos , Humanos , Indio/química , Ratones , Imagen Óptica , Pseudomonas aeruginosa/efectos de los fármacos , Esterilización , Sulfuros/química , Propiedades de Superficie , Compuestos de Zinc/química
13.
J Mater Chem B ; 9(5): 1364-1369, 2021 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-33458729

RESUMEN

Obstinate infections caused by drug-resistant bacteria severely threaten human health. And the emergence of multidrug-resistant bacteria increases the morbidity and mortality of patients, thus necessitating the development of innovative or alternative therapeutics. Here, a light-activated nanotherapeutic with broad-spectrum bacterial recognition is established as an antibiotic-free therapeutic agent against pathogens. The nanotherapeutic with external phenylboronic acid-based glycopolymers increases the stability and biocompatibility and shows the ability of bacterial recognition. Once irradiated with near-infrared light, this nanotherapeutic with high photothermal conversion efficiency disrupts the cytoplasmic membrane, thus killing bacterial cells. Importantly, it also eliminates the biofilms formed by both drug-resistant Gram-negative bacteria (Pseudomonas aeruginosa) and Gram-positive bacteria (Staphylococcus aureus) effectively. Thus, this antibiotic-free nanotherapeutic with hypotoxicity offers a promising approach to fight increasingly serious antimicrobial resistance.


Asunto(s)
Antibacterianos/uso terapéutico , Farmacorresistencia Bacteriana Múltiple/genética , Bacterias Grampositivas/patogenicidad , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Nanotecnología/métodos , Antibacterianos/farmacología , Humanos
14.
Biomater Sci ; 8(24): 6912-6919, 2020 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-32749389

RESUMEN

The Gram-negative opportunistic pathogen Pseudomonas aeruginosa is endowed with intrinsic resistance to antibiotics. It is essential to explore alternative techniques to supplement the arsenal of methods to kill drug-resistant bacteria. Herein, we established an "on-demand" nanoplatform based on acid-degradable scaffolds by conjugating glycomimetic-based galactose ligands to target a key lectin on P. aeruginosa and guanidine moieties. This nanoplatform could capture bacteria through ligand-receptor interactions and electrostatic interactions, and subsequently reactive oxygen species produced by entrapped photodynamic agent Ce6 under light irradiation eliminated drug-resistant P. aeruginosa and its biofilm. Approximately 95% of the planktonic bacteria were killed and more than 70% of the biofilm was disrupted under light irradiation. This strategy of copolymer modification could improve the biocompatibility and therapeutic efficiency levels of antibacterial therapeutics through the targeting of function. Hence, utilizing this smart nanoplatform may be of significance in developing new strategies to solve the growing problem of bacterial resistance.


Asunto(s)
Preparaciones Farmacéuticas , Fotoquimioterapia , Antibacterianos/farmacología , Biopelículas , Pseudomonas aeruginosa
15.
Front Pharmacol ; 11: 164, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32194406

RESUMEN

In this study, we found miR-362-5p was upregulated in bladder cancer tissues and we predicted that QKI is potential a target of miR-362-5p and MBNL1-AS1 might be able to directly target to miR-362-5p. We attempted to evaluate whether miR-362-5p could play its roles in bladder cancer through regulating QKI (quaking) and whether the expression and function of miR-362-5p could be mediated by lncRNA MBNL1-AS1. We performed the gain- and loss-function experiments to explore the association between miR-362-5p expression and bladder cancer proliferation. In vivo, the nude mice were injected with miR-362-5p knockdown SW780 cells to assess the effects of miR-362-5p on tumor growth. The results showed upregulation of miR-362-5p promoted cell proliferation of bladder cancer cells. MBNL1-AS1 and QKI could directly bind with miR-362-5p, and knockdown of MBNL1-AS1 or QKI could abrogate the regulatory effects of miR-362-5p on bladder cancer cell proliferation. Furthermore, downregulation of miR-362-5p inhibited bladder tumor growth and increased QKI expression. Our data unveiled that miR-362-5p may play an oncogenic role in bladder cancer through QKI and MBNL1-AS1 might function as a sponge to mediate the miR-362-5p expression and function.

16.
ACS Appl Bio Mater ; 3(4): 2132-2139, 2020 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-35025265

RESUMEN

Insulin injection causes great pain to the patient, and nasal mucosal administration of insulin is a novel route for the treatment of diabetes. This strategy could protect insulin from either extensive first-pass metabolism or enzyme degradation in the gastrointestinal tract. With the dynamic boronate esters reversibly formed by phenylboronic acid and diols on nasal mucosal epithelial cell surfaces, we herein developed phenylboronic-acid-functionalized dextran nanoplatforms to enhance the permeability of cargos and boost penetration. The nanoplatforms with excellent loading capacity exhibited significant endocytosis compared with naked insulin. The mechanism of endocytosis was involved in clathrin- and lipid raft/caveolae-dependent endocytic pathways. The in vivo nasal delivery of insulin suggested that these nanoplatforms did not trigger nasal epithelial inflammation and greatly decreased blood sugar levels and improved insulin bioavailability. Collectively, this proof-of-concept study demonstrates a novel carrier of phenylboronic-acid-decorated polymer for insulin delivery and provides a promising approach for the development of a diabetes therapeutic strategy.

17.
Cancer Med ; 9(2): 724-736, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31769229

RESUMEN

LncRNAs have been shown to play essential roles in bladder cancer (BC) progress. Our microarrays of clinical samples firstly screened that lncRNA muscleblind-like 1 antisense RNA 1 (MBNL1-AS1) was poorly expressed in BC tissues. However, its biological function in BC remains not well understood. Here we examined the clinical correlations with MBNL1-AS1 in BC patients. Then, 5673 and T24 cell lines were employed to investigate the role of MBNL1-AS1 in the proliferation and apoptosis of BC cells in vitro and in vivo. Furthermore, miR-135a-5p (miR-135a)/PHLPP2/FOXO1 axis was focused to explore its regulatory mechanism in BC. The results showed that MBNL1-AS1 was significantly downregulated in bladder tumor tissues, and associated with BC progression. In vitro, MBNL1-AS1 knockdown increased the number of viable cells and bromodeoxyuridine-positive cells, accelerated cell cycle, and dysregulated proliferative regulators (Ki67, p21, p27, and Cyclin D1) in BC cells. The apoptotic cells and the cleavages of caspase-3/9 were reduced in MBNL1-AS1-silenced BC cells. Overexpression of MBNL1-AS1 had opposite effects on BC cell proliferation and apoptosis. Moreover miR-135a was demonstrated to interact with MBNL1-AS1, and inhibiting miR-135a reversed the effects of shMBNL1-AS1 on BC cells. The downstream effectors (PHLPP2 and FOXO1) were positively regulated by MBNL1-AS1, but negatively regulated by miR-135a. Similar results were also observed in xenograft tumors. In conclusion, this study firstly suggests that MBNL1-AS1 acts as a tumor suppressor of BC by targeting miR-135a/PHLPP2/FOXO1 axis, providing a novel insight for BC diagnosis and treatment.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Proteína Forkhead Box O1/metabolismo , MicroARNs/genética , Fosfoproteínas Fosfatasas/metabolismo , ARN Largo no Codificante/genética , Proteínas de Unión al ARN/antagonistas & inhibidores , Neoplasias de la Vejiga Urinaria/patología , Apoptosis , Biomarcadores de Tumor/genética , Proliferación Celular , Femenino , Proteína Forkhead Box O1/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Fosfoproteínas Fosfatasas/genética , Pronóstico , ARN sin Sentido/genética , Proteínas de Unión al ARN/genética , Células Tumorales Cultivadas , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/metabolismo
18.
ACS Appl Mater Interfaces ; 11(43): 39648-39661, 2019 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-31591880

RESUMEN

Pseudomonas aeruginosa can cause a multitude of inflammations in humans. Due to its ability to form biofilm, the bacteria show durable resistance to drugs. Herein, we developed a heteromultivalent ligand-decorated nanotherapeutic inspired by living system for inhibition of antibiotic-resistant bacterial pneumonia. The nanotherapeutic with a heteromultivalent glycomimetic shell can specifically recognize P. aeruginosa to inhibit its biofilm formation and protect native cells from bacterial infection; the rate of biofilm inhibition was up to 85%. The nanotherapeutic with a bioresponsive hydrophobic core can protonate and control drug release in the microenvironment of bacterial infections. By utilizing these properties, the nanotherapeutics can effectively penetrate the internal structure of biofilms to release the drug, dispersing the biofilm by over 80% under laser irradiation. In vivo bioinspired nanotherapeutics have the potential to efficiently inhibit antibiotic-resistant P. aeruginosa-induced pneumonia. Collectively, we expect biomimicking systems to be the next generation of prevention and treatment as integrated antibacterial agents against P. aeruginosa.


Asunto(s)
Biopelículas/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Farmacorresistencia Bacteriana/efectos de los fármacos , Nanopartículas , Neumonía Bacteriana/tratamiento farmacológico , Infecciones por Pseudomonas/tratamiento farmacológico , Pseudomonas aeruginosa/fisiología , Células A549 , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Nanopartículas/química , Nanopartículas/uso terapéutico , Fotoquimioterapia , Neumonía Bacteriana/metabolismo , Neumonía Bacteriana/patología , Infecciones por Pseudomonas/metabolismo , Infecciones por Pseudomonas/patología , Nanomedicina Teranóstica
19.
ACS Appl Bio Mater ; 2(4): 1614-1622, 2019 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-35026895

RESUMEN

The Gram-negative bacteria Pseudomonas aeruginosa is one famous bacterial strain owing to its ability to effectively form biofilms, which is a front-line mechanism of bacterial tolerance. Herein, the near-infrared-induced nanocomposites were one-step prepared by modifying copper sulfide nanoparticle with peptide to effectively eradicate Pseudomonas aeruginosa biofilm through electrostatic interaction, photodynamic effect and photothermal effect. These nanocomposites could rapidly adhere to the surface of bacteria, and irreversible damage the bacterial membrane under near-infrared laser irradiation. Furthermore, the nanocomposites could selectively eliminate bacteria over mammalian cell without distinct toxicity to NIH 3T3 cells. The nanocomposites will exert a far-reaching impact on the future design of biocompatible near-infrared-induced antibacterial agents, exhibiting its potential applications in Gram-negative bacteria and biofilm infections.

20.
Adv Mater ; 31(7): e1806024, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30589118

RESUMEN

The chronic infections by pathogenic Pseudomonas aeruginosa (P. aeruginosa) remain to be properly addressed. In particular, for drug-resistant strains, limited medication is available. An in vivo pneumonia model induced by a clinically isolated aminoglycoside resistant strain of P. aeruginosa is developed. Tobramycin clinically treating P. aeruginosa infections is found to be ineffective to inhibit or eliminate this drug-resistant strain. Here, a newly developed non-antibiotics based nanoformulation plus near-infrared (NIR) photothermal treatment shows a remarkable antibacterial efficacy in treating this drug-resistant pneumonia. The novel formulation contains 50-100 nm long nanorods decorated with two types of glycomimetic polymers to specifically block bacterial LecA and LecB lectins, respectively, which are essential for bacterial biofilm development. Such a 3D display of heteromultivalent glycomimetics on a large scale is inspired by the natural strengthening mechanism for the carbohydrate-lectin interaction that occurs when bacteria initially infects the host. This novel formulation shows the most efficient bacteria inhabitation and killing against P. aeruginosa infection, through lectin blocking and the near-infrared-light-induced photothermal effect of gold nanorods, respectively. Collectively, the novel biomimetic design combined with the photothermal killing capability is expected to be an alternative treatment strategy against the ever-threatening drug-resistant infectious diseases when known antibiotics have failed.


Asunto(s)
Materiales Biomiméticos , Hipertermia Inducida/métodos , Fototerapia/métodos , Infecciones por Pseudomonas/terapia , Pseudomonas aeruginosa , Células A549 , Absceso/tratamiento farmacológico , Absceso/patología , Adhesinas Bacterianas/metabolismo , Animales , Biopelículas , Farmacorresistencia Bacteriana , Escherichia coli , Compuestos de Oro , Humanos , Lactosa/análogos & derivados , Lectinas/antagonistas & inhibidores , Lectinas/metabolismo , Ratones , Ratones Endogámicos BALB C , Células 3T3 NIH , Nanotubos , Neumonía Bacteriana/patología , Neumonía Bacteriana/terapia , Ácidos Polimetacrílicos , Infecciones por Pseudomonas/patología , Pseudomonas aeruginosa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...