Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
J Hematol Oncol ; 17(1): 23, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38659046

RESUMEN

BACKGROUND: The autologous anti-B-cell maturation antigen (BCMA) chimeric antigen receptor (CAR) T-cell therapy LCAR-B38M has been approved for the treatment of relapsed and refractory multiple myeloma in many countries across the world under the name ciltacabtagene autoleucel. LEGEND-2 was the first-in-human trial of LCAR-B38M and yielded deep and durable therapeutic responses. Here, we reported the outcomes in LEGEND-2 after a minimal 5-year follow-up. METHODS: Participants received an average dose of 0.5 × 106 cells/kg LCAR-B38M in split or single unfractionated infusions after cyclophosphamide-based lymphodepletion therapy. Investigator-assessed response, survival, safety and pharmacokinetics were evaluated. RESULTS: Seventy-four participants enrolled and had a median follow-up of 65.4 months. The 5-year progression-free survival (PFS) and overall survival (OS) rates were 21.0% and 49.1%, with progressive flattening of the survival curves over time. Patients with complete response (CR) had longer PFS and OS, with 5-year rates of 28.4% and 65.7%, respectively. Twelve patients (16.2%) remained relapse-free irrespective of baseline high-risk cytogenetic abnormality and all had normal humoral immunity reconstituted. An ongoing CR closely correlated with several prognostic baseline indices including favorable performance status, immunoglobulin G subtype, and absence of extramedullary disease, as well as a combination cyclophosphamide and fludarabine preconditioning strategy. Sixty-two (83.8%) suffered progressive disease (PD) and/or death; however, 61.1% of PD patients could well respond to subsequent therapies, among which, the proteasome inhibitor-based regimens benefited the most. Concerning the safety, hematologic and hepatic function recovery were not significantly different between non-PD and PD/Death groups. A low rate of second primary malignancy (5.4%) and no severe virus infection were observed. The patients who tested positive for COVID-19 merely presented self-limiting symptoms. In addition, a sustainable CAR T population of one case with persistent remission was delineated, which was enriched with indolently proliferative and lowly cytotoxic CD4/CD8 double-negative functional T lymphocytes. CONCLUSIONS: These data, representing the longest follow-up of BCMA-redirected CAR T-cell therapy to date, demonstrate long-term remission and survival with LCAR-B38M for advanced myeloma. TRIAL REGISTRATION: LEGEND-2 was registered under the trial numbers NCT03090659, ChiCTRONH-17012285.


Asunto(s)
Antígeno de Maduración de Linfocitos B , Inmunoterapia Adoptiva , Mieloma Múltiple , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Antígeno de Maduración de Linfocitos B/inmunología , Estudios de Seguimiento , Inmunoterapia Adoptiva/métodos , Inmunoterapia Adoptiva/efectos adversos , Mieloma Múltiple/terapia , Mieloma Múltiple/mortalidad , Receptores Quiméricos de Antígenos/uso terapéutico , Receptores Quiméricos de Antígenos/inmunología , Inducción de Remisión , Tasa de Supervivencia
2.
Drug Resist Updat ; 73: 101059, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38295753

RESUMEN

Patients with bladder cancer (BCa) frequently acquires resistance to platinum-based chemotherapy, particularly cisplatin. This study centered on the mechanism of cisplatin resistance in BCa and highlighted the pivotal role of lactylation in driving this phenomenon. Utilizing single-cell RNA sequencing, we delineated the single-cell landscape of Bca, pinpointing a distinctive subset of BCa cells that exhibit marked resistance to cisplatin with association with glycolysis metabolism. Notably, we observed that H3 lysine 18 lactylation (H3K18la) plays a crucial role in activating the transcription of target genes by enriching in their promoter regions. Targeted inhibition of H3K18la effectively restored cisplatin sensitivity in these cisplatin-resistant epithelial cells. Furthermore, H3K18la-driven key transcription factors YBX1 and YY1 promote cisplatin resistance in BCa. These findings enhance our understanding of the mechanisms underlying cisplatin resistance, offering valuable insights for identifying novel intervention targets to overcome drug resistance in Bca.


Asunto(s)
Cisplatino , Neoplasias de la Vejiga Urinaria , Humanos , Cisplatino/farmacología , Cisplatino/uso terapéutico , Histonas/genética , Histonas/metabolismo , Análisis de Expresión Génica de una Sola Célula , Resistencia a Antineoplásicos/genética , Línea Celular Tumoral , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/metabolismo
3.
Anal Chim Acta ; 1272: 341482, 2023 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-37355329

RESUMEN

ß-galactosidase (ß-Gal) is an important biomarker of cell senescence and primary ovarian cancer. Therefore, it is of great significance to construct a near-infrared fluorescent probe with deep tissue penetration and a high signal-to-noise ratio for visualization of ß-galactosidase in biological systems. However, most near-infrared probes tend to have small Stokes shifts and low signal-to-noise ratios due to crosstalk between excitation and emission spectra. Using d-galactose residues as specific recognition units and near-infrared dye TJ730 as fluorophores, a near-infrared fluorescence probe SN-CR with asymmetric structure was developed for the detection of ß-Gal. The probe has a fast reaction equilibrium time (<12 min) with ß-Gal, excellent biocompatibility, near-infrared emission (738 nm), low detection limit (0.0029 U/mL), and no crosstalk between the excitation spectrum and emission spectrum (Stokes shifts 142 nm) of the probe. Cell imaging studies have shown that SN-CR can visually trace ß-Gal in different cells and distinguish ovarian cancer cells from other cells.


Asunto(s)
Sondas Moleculares , beta-Galactosidasa , Células HeLa , Línea Celular , Humanos , Animales , Perros , beta-Galactosidasa/análisis , Sondas Moleculares/síntesis química , Sondas Moleculares/química , Fluorescencia
4.
Biosens Bioelectron ; 236: 115401, 2023 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-37257317

RESUMEN

Ovarian cancer is a gynecologic malignancy with high mortality. The main reason is that it is detected at an advanced stage due to a lack of early diagnosis and treatment. Therefore, it is of great interest to develop a chemical tool that can visualize ovarian cancer cells in real-time and eliminate them. Unfortunately, probes that can simultaneously monitor both modes of action for the diagnosis and treatment of ovarian cancer have not been developed. Here, we designed a novel prodrug fluorescent probe (YW-OAc) that not only visually tracks cancer cells but also enables the on-demand delivery of chemotherapeutic agents. By ß-Gal-mediated glycosidic bond hydrolysis, the fluorescent signal changed from blue to green (signal 1), enabling visual tracking of ovarian cancer cells. Subsequently, the identified cancer cells were subjected to precise light irradiation to induce anticancer drug release accompanied by a fluorescence transition from green to blue (signal 2), enabling real-time information on drug release. Thus, the prodrug fluorescent probe YW-OAc provides comprehensive two-step monitoring during cancer cell recognition and clearance. Notably, YW-OAc exhibited high affinity (Km = 3.74 µM), high selectivity, and low detection limit for ß-Gal (0.0035 U/mL). We also demonstrated that YW-OAc can visually trace endogenous ß-Gal in different cells and exhibit high phototoxicity in ovarian cancer cells. We hope that the prodrug fluorescent probe YW-OAc, can be used as an effective tool for biomedical diagnosis and treatment.


Asunto(s)
Antineoplásicos , Técnicas Biosensibles , Neoplasias Ováricas , Profármacos , Femenino , Humanos , Profármacos/farmacología , Profármacos/química , Colorantes Fluorescentes/química , Antineoplásicos/farmacología , Antineoplásicos/química , Neoplasias Ováricas/diagnóstico , Neoplasias Ováricas/tratamiento farmacológico , beta-Galactosidasa
5.
J Med Virol ; 95(2): e28501, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36655747

RESUMEN

Data on the dynamic changes in chronic hepatitis B (CHB) patients with nonalcoholic fatty liver disease (NAFLD) during antiviral therapy are scarce. We aimed to investigate the evolution of NAFLD status change in CHB patients treated with nucleos(t)ide analogues (NAs) and its influence on therapeutic outcomes. This retrospective study included 164 HBeAg-positive CHB patients from a randomized controlled trial who were treated with NAs for 104 weeks and underwent paired liver biopsies. Histological evaluation was performed at baseline and Week 104. The patients were divided into four groups according to NAFLD status changes. From baseline to Week 104, the overall percentage of CHB patients with concurrent NAFLD increased from 17.1% to 26.2% (p = 0.044). Among them, 7 of 28 patients (25.0%) with NAFLD at baseline showed NAFLD remission at week 104, while 22 of 136 patients (16.2%) without NAFLD at baseline developed new-onset NAFLD. In subgroup analyses, the new-onset and sustained NAFLD groups showed significantly lower rates of biochemical response at week 104 as compared to the sustained non-NAFLD group (77.3% and 57.1% vs. 93.9%, respectively; all p < 0.05), as well as fibrosis improvement (31.8% and 42.9% vs. 69.3%, respectively; all p < 0.05). NAFLD status changes did not influence the virological response, HBeAg seroconversion, and necroinflammation improvement (all p > 0.05). In HBeAg-positive CHB patients receiving NAs therapy, new-onset and sustained NAFLD may counteract the benefits of antiviral therapy, reducing the rate of biochemical response and fibrosis improvement.


Asunto(s)
Hepatitis B Crónica , Enfermedad del Hígado Graso no Alcohólico , Humanos , Antivirales/uso terapéutico , Antígenos e de la Hepatitis B/análisis , Hepatitis B Crónica/tratamiento farmacológico , Resultado del Tratamiento , Estudios Retrospectivos , Fibrosis , Virus de la Hepatitis B
6.
J Chemother ; 35(1): 11-18, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34927575

RESUMEN

Sunitinib, an inhibitor of receptor tyrosine kinase, possesses anti-tumor activity in renal cell carcinoma (RCC) through its anti-angiogenic effects. However, patients with advanced RCC are resistant to sunitinib. Dysregulated circRNAs has been shown to be associated with drug resistance in various tumors. However, little is known about the effect of circRNA_001895 on sunitinib resistance of RCC. First, the expression of circRNA_001895 was found to be higher in sunitinib-resistant RCC tissues than chemosensitive tumor tissues. Half maximal inhibitory concentration of sunitinib-resistant RCC cells (786-O/R and ACHN/R) was higher than sunitinib-sensitive 786-O and ACHN cells. CircRNA_001895 was also upregulated in 786-O/R and ACHN/R cells. Second, data from colony formation and flow cytometry analysis showed that knockdown of circRNA_001895 suppressed cell proliferation and promoted cell apoptosis in 786-O/R and ACHN/R cells. Moreover, the protein expression of phosphorylated histone H2AX (γH2AX) was enhanced, while phosphorylated DNA-dependent protein kinase (p-DNA-PK) and Rad51 were reduced in 786-/R and ACHN/R by knockdown of circRNA_001895. Lastly, knockdown of circRNA_001895 conferred sensitivity of in vivo tumor growth to sunitinib. In conclusion, circRNA_001895 was implicated in the sunitinib resistance in RCC through regulation of apoptosis and DNA damage repair, suggesting that circRNA_001895 might be a potential therapeutic target for advanced RCC.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Humanos , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Sunitinib/farmacología , Sunitinib/uso terapéutico , ARN Circular/genética , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/genética , Neoplasias Renales/patología , Resistencia a Antineoplásicos/genética , Proliferación Celular , Apoptosis , Daño del ADN , Línea Celular Tumoral
7.
Spectrochim Acta A Mol Biomol Spectrosc ; 285: 121879, 2023 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-36122464

RESUMEN

ß-Galactosidase (ß-gal) is a hydrolytic enzyme in lysosomes and is also an important biomarker of cellular senescence and primary ovarian cancer. Therefore, real-time non-invasive detection of ß-gal activity in vivo is of great significance for the prevention of cell senescence and early diagnosis of ovarian cancer. We designed an enzyme-activated proportional near-infrared (NIR) probe (Gal-Br-NO2) for real-time fluorescence quantification and capture of ß-gal activity in vivo. The main characteristics of the Gal-Br-NO2 probe include short response time (less than 10 min), large Stokes displacement (155 nm), and near-infrared fluorescence emission (670 nm). The probe has also been successfully used to detect ß-gal in ovarian cancer cells and senile cells and can accurately detect endogenous ß-gal in zebrafish. Our work provides a potential tool for pre-clinical real-time tracking of ß-gal activity in vivo and early diagnosis of disease.


Asunto(s)
Colorantes Fluorescentes , Neoplasias Ováricas , Animales , Femenino , Humanos , Pez Cebra , Dióxido de Nitrógeno , beta-Galactosidasa
8.
Anal Chim Acta ; 1232: 340459, 2022 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-36257743

RESUMEN

Overexpression of ß-galactosidase (ß-gal) in tumor cells may serve as a valuable biomarker for the early diagnosis of some cancers (such as ovarian cancer). In addition, abnormal accumulation of ß-gal is also considered an essential marker of cell senescence. Therefore, it is important to construct fluorescent probes with excellent fluorescence properties to visualize ß-gal in biological systems. Here, we designed and screened a novel fluorescent probe XM for the detection of ß-gal. Spectral data show that the probe has a good affinity (Km = 2.6 µM) for ß-gal, large stokes shift (190 nm), fast response speed (stable within 20 min), and low detection limit (6.7 × 10-3 U/mL). Based on the above advantages, XM can not only detect ß-gal content in cancer cells but also track the changes of ß-gal content in zebrafish at different developmental period. We believe XM will become a powerful tool for the early cancer diagnosis and cellular senescence.


Asunto(s)
Colorantes Fluorescentes , Pez Cebra , Animales , beta-Galactosidasa , Microscopía Fluorescente
9.
Theriogenology ; 189: 301-312, 2022 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-35842953

RESUMEN

Orchitis accounts for a high proportion of male animal reproductive disorders. Hence, it is urgent to identify drugs for the prevention and treatment of orchitis. Antimicrobial peptides (AMPs) are currently recognized as one of the most promising alternatives to antibiotics. However, the protective effects of AMPs on lipopolysaccharide (LPS)-induced orchitis have not been reported. In this study, we developed an LPS-induced orchitis model in which primary bovine Sertoli cells were used as model cells. MPX was indicated to effectively reduce the inflammatory response of Sertoli cells. MPX attenuated the gene expression of the proinflammatory cytokines TNF-α, IL-6 and IL-1ß by suppressing the MAPK pathway, especially the phosphorylation of p38 and ERK. MPX also decreased the oxidative stress response caused by LPS and upregulated Occludin and Claudin-1 expression, thereby maintaining the integrity of the blood-testis barrier. Moreover, we found that MPX inhibited apoptosis in Sertoli cells. In a mouse model, we found that MPX significantly inhibited the disruptive effects of LPS, reducing seminiferous epithelium damage, vacuolations, hyperplasia, and apoptosis in spermatogenic cells and rescuing spermatogenesis. In addition, the expression of inflammatory factors such as IL-1ß, IL-18, IL-6 and TNF-α was decreased after MPX treatment in the mouse testes. MPX had no effect on other organs in mice, indicating its safety. This study was undertaken to investigate how MPX regulates the inflammatory response in Sertoli cells and provide a reference for the clinical prevention and treatment of male animal orchitis.


Asunto(s)
Enfermedades de los Bovinos , Orquitis , Enfermedades de los Roedores , Animales , Péptidos Antimicrobianos , Barrera Hematotesticular/metabolismo , Bovinos , Enfermedades de los Bovinos/metabolismo , Interleucina-6/metabolismo , Lipopolisacáridos/toxicidad , Masculino , Ratones , Orquitis/tratamiento farmacológico , Orquitis/metabolismo , Orquitis/veterinaria , Enfermedades de los Roedores/metabolismo , Células de Sertoli/metabolismo , Testículo , Factor de Necrosis Tumoral alfa/metabolismo
10.
Rev. Assoc. Med. Bras. (1992, Impr.) ; Rev. Assoc. Med. Bras. (1992, Impr.);67(9): 1293-1298, Sept. 2021. tab
Artículo en Inglés | LILACS | ID: biblio-1351485

RESUMEN

SUMMARY OBJECTIVE: Our study aimed to explore the potential risk factors for radiological hip joint involvement in patients with ankylosing spondylitis (AS). METHODS: This cross-sectional convey collected the clinical data, laboratory indicators, and radiographic data of patients with AS. Radiographic hip joint involvement was defined as a Bath Ankylosing Spondylitis Radiology Hip Index (BASRI-hip) score ≥2. Multivariate logistic regression analyses were conducted to explore the potential risk factors for radiological hip involvement in patients with AS. RESULTS: Based on BASRI-hip score, all enrolled 386 patients with AS were classified as patients involving with radiological hip joint involvement (BASRI-hip ≥2; n=203) and those without it (BASRI-hip ≤1; n=183). Mean age of enrolled patients with AS were 36.7±11.9 years, and 320 (82.9%) patients were male. Mean course of disease was 10.7±8.3 years, and 349 (90.4%) patients were with a positive HLAB27. Multivariate analyses indicated that Juvenile onset (onset age ≤16 years) (odds ratio [OR]=4.159, 95% confidence interval [CI], 1.779-9.721, p<0.001), body mass index (BMI) <18.5 kg/m2 (OR=1.986, 95%CI 1.187-3.323, p=0.009), continuous nonsteroidal anti-inflammatory drug (NSAID) use (OR=0.351, 95%CI 0.155-0.794, p=0.012), and bone mass below the expected range for age (Z score ≤-2) (OR=2.791, 95%CI 1.456-5.352, p=0.002) were independently associated with radiological hip joint involvement in patients with AS. CONCLUSIONS: The potential risk factors for radiological hip joint involvement were juvenile onset, lower BMI, and bone mass below the expected range for age. Furthermore, continuous NSAID use was the protective factor for radiological hip joint involvement in these population.


Asunto(s)
Humanos , Masculino , Adulto , Espondilitis Anquilosante/complicaciones , Espondilitis Anquilosante/diagnóstico por imagen , Articulación de la Cadera/fisiopatología , Índice de Severidad de la Enfermedad , Índice de Masa Corporal , Densidad Ósea , Antiinflamatorios no Esteroideos/uso terapéutico , Estudios Transversales , Factores de Riesgo , Edad de Inicio , Articulación de la Cadera/diagnóstico por imagen , Persona de Mediana Edad
11.
Front Microbiol ; 12: 644887, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34177825

RESUMEN

Escherichia coli can cause intestinal diseases in humans and livestock, destroy the intestinal barrier, exacerbate systemic inflammation, and seriously threaten human health and animal husbandry development. The aim of this study was to investigate whether the antimicrobial peptide mastoparan X (MPX) was effective against E. coli infection. BALB/c mice infected with E. coli by intraperitoneal injection, which represents a sepsis model. In this study, MPX exhibited no toxicity in IPEC-J2 cells and notably suppressed the levels of interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), myeloperoxidase (MPO), and lactate dehydrogenase (LDH) released by E. coli. In addition, MPX improved the expression of ZO-1, occludin, and claudin and enhanced the wound healing of IPEC-J2 cells. The therapeutic effect of MPX was evaluated in a murine model, revealing that it protected mice from lethal E. coli infection. Furthermore, MPX increased the length of villi and reduced the infiltration of inflammatory cells into the jejunum. SEM and TEM analyses showed that MPX effectively ameliorated the jejunum damage caused by E. coli and increased the number and length of microvilli. In addition, MPX decreased the expression of IL-2, IL-6, TNF-α, p-p38, and p-p65 in the jejunum and colon. Moreover, MPX increased the expression of ZO-1, occludin, and MUC2 in the jejunum and colon, improved the function of the intestinal barrier, and promoted the absorption of nutrients. This study suggests that MPX is an effective therapeutic agent for E. coli infection and other intestinal diseases, laying the foundation for the development of new drugs for bacterial infections.

12.
Mol Med Rep ; 23(1)2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33236143

RESUMEN

Macrophage­capping protein (CapG) is a newly characterized oncogene involved in several types of cancer. However, the expression patterns and biological mechanisms of CapG in clear cell renal cell carcinoma (ccRCC) are unclear. The present study aimed to investigate the roles of CapG in the prognosis, proliferation and metastasis of ccRCC. In the present study, the expression of CapG was analyzed by western blotting in 24 paired ccRCC and adjacent normal tissue samples. Another 152 tissue samples from 152 patients with ccRCC were examined by immunohistochemistry. Compared with normal tissue, CapG expression was significantly increased in ccRCC tissue, and high CapG expression was associated with advanced tumor stage, histological grade, lymph node metastasis, and poor overall survival. Moreover, CapG was an independent predictor of survival. Lentivirus­mediated CapG knockdown significantly inhibited 786­O cell proliferation, migration, and invasion, induced cell cycle arrest at the G2/M phase, and increased apoptosis in vitro. Microarray analysis indicated that RAC, CDC42 and ERK/MAPK signaling were disrupted by CapG knockdown in 786­O cells. In conclusion, the present findings indicate that CapG plays an oncogenic role in ccRCC and may represent a potential therapeutic target for this disease.


Asunto(s)
Carcinoma de Células Renales/metabolismo , Neoplasias Renales/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Oncogénicas/metabolismo , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/mortalidad , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Movimiento Celular , Puntos de Control de la Fase G2 del Ciclo Celular , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Renales/genética , Neoplasias Renales/mortalidad , Neoplasias Renales/patología , Puntos de Control de la Fase M del Ciclo Celular , Sistema de Señalización de MAP Quinasas , Proteínas de Microfilamentos/genética , Proteínas Nucleares/genética , Proteínas Oncogénicas/genética
13.
Food Chem ; 324: 126888, 2020 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-32344352

RESUMEN

This study aimed to investigate if myo-inositol (MI) supplementation could alleviate adverse effects caused by aflatoxin B1 (AFB1) with respect to growth performance, AFB1 residues, immune response and antioxidant status of Litopenaeus vannamei. 800 shrimp (initial weight: 1.1 g) were divided into five groups: MI0 (basal diet); MI0 + LA, MI0 + HA, MI200 + LA and MI200 + HA fed with AFB1-contaminated diets (LA, low concentration AFB1; HA, high concentration AFB1; MI200, adding 200 mg MI kg-1 diet). The results showed that HA significantly decreased growth performance, systemic inositol content and lipid content. AFB1 residues were detected in the hepatopancreas of shrimp, but not the muscle. Histological lesions were observed in MI0 + LA and MI0 + HA groups. HA supplementation raised malondialdehyde and protein carbonyl content and reduced some antioxidant enzyme activities and immune-related genes expression, which was slightly ameliorated by MI supplementation. Our results suggest that myo-inositol may slightly mitigate negative impacts caused by AFB1 in L. vannamei.


Asunto(s)
Aflatoxina B1/análisis , Antioxidantes/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Inositol/farmacología , Penaeidae/crecimiento & desarrollo , Aflatoxina B1/administración & dosificación , Alanina Transaminasa/metabolismo , Animales , Aspartato Aminotransferasas/metabolismo , Dieta , Suplementos Dietéticos , Hepatopáncreas/enzimología , Hepatopáncreas/metabolismo , Malondialdehído/metabolismo , Penaeidae/inmunología , Penaeidae/metabolismo , Carbonilación Proteica/efectos de los fármacos
14.
Exp Cell Res ; 391(1): 112004, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32289284

RESUMEN

Deubiquitinase (DUB) can reverse the ubiquitin signal, and participate in virtually all aspects of cancer progression. Thus, DUB represents an attractive target for development of anticancer drugs. However, little is known about DUB which can be used as drug targets. Here, we found that the constitutive photomorphogenic 9 (COP9) signalosome complex subunit 6 (COPS6/CSN6), a DUB belongs to JAMM/MPN domain-associated metallopeptidases(JAMMs) class, was highly expressed in pancreatic adenocarcinoma(PAAD) tissues. High expression of CSN6 was associated with tumor TNM stage and metastasis in PAAD patients. Moreover, we demonstrated that CSN6 promoted invasion and metastasis through regulating forkhead box protein A1 (FOXA1) in PAAD cells. Re-expression of FOXA1 rescued the decreased invasion and metastasis caused by CSN6 knockdown, whereas inhibition of FOXA1 alleviated the pro-metastasis effect induced by CSN6 overexpression. Further, CSN6 regulated the expression of FOXA1 via c-Fos in PAAD cells. Mechanistically, CSN6 stabilized c-Fos protein by binding to it and decreasing its ubiquitination. Our work identified CSN6 as a targeting-permissible deubiquitinase, and CSN6 inhibition maybe a potential treatment strategy for PAAD.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Adenocarcinoma/genética , Complejo del Señalosoma COP9/genética , Regulación Neoplásica de la Expresión Génica , Factor Nuclear 3-alfa del Hepatocito/genética , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/mortalidad , Adenocarcinoma/patología , Anciano , Complejo del Señalosoma COP9/antagonistas & inhibidores , Complejo del Señalosoma COP9/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Factor Nuclear 3-alfa del Hepatocito/antagonistas & inhibidores , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Humanos , Leupeptinas/farmacología , Metástasis Linfática , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Análisis de Supervivencia , Ubiquitina/genética , Ubiquitina/metabolismo , Ubiquitinación , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Cancer Sci ; 111(2): 713-726, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31782868

RESUMEN

There is an urgent need to find novel potential therapeutic targets for the diagnosis and treatment of clear cell renal cell carcinoma (ccRCC) due to its highly invasive ability as a common urological malignant tumor. Circular RNAs (circRNAs) have been indicated as potentially critical mediators in various types of tumor progression. We first used qRT-PCR analysis to find dysregulated circRNAs in ccRCC. A novel circRNA, hsa_circ_001895, was upregulated in ccRCC specimens and associated with metastatic properties of ccRCC. However, the tumorigenic mechanism of hsa_circ_001895 on ccRCC is yet to be found. We first indicated that hsa_circ_001895 predicted a poor prognosis in ccRCC patients. Additionally, overexpression of hsa_circ_001895 not only promoted cell proliferation, invasion and migration of ccRCC, but also inhibited cell apoptosis, whereas hsa_circ_001895 knockdown reversed the effect on ccRCC progression. In vivo s.c. xenotransplanted tumor model also showed that silencing hsa_circ_001895 could suppress in vivo ccRCC growth. Mechanistically, hsa_circ_001895 directly binds with microRNA (miR)-296-5p and inhibits its expression. Moreover, sex determining region Y (SRY)-box 12 (SOX12) was identified as a target of miR-296-5p, the expression of which was suppressed by miR-296-5p. Notably, the inhibitory effect of hsa_circ_001895 on ccRCC progression was reversed by miR-296-5p inhibitor. In general, our findings indicated that hsa_circ_001895 may sponge miR-296-5p and promote SOX12 expression, which is the underlying mechanism of hsa_circ_001895-induced ccRCC progression.


Asunto(s)
Carcinoma de Células Renales/patología , Neoplasias Renales/patología , MicroARNs/genética , ARN Circular/genética , Factores de Transcripción SOXC/genética , Regiones no Traducidas 3' , Animales , Biomarcadores de Tumor/genética , Carcinoma de Células Renales/genética , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Renales/genética , Masculino , Ratones , Metástasis de la Neoplasia , Estadificación de Neoplasias , Trasplante de Neoplasias , Pronóstico
16.
Clin Gastroenterol Hepatol ; 18(2): 457-467.e21, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31306800

RESUMEN

BACKGROUND & AIMS: Treatment of chronic hepatitis B virus (HBV) infection with entecavir suppresses virus replication and reduces disease progression, but could require life-long therapy. To investigate clinical outcome events and safety associated with long-term treatment with entecavir, we followed up patients treated with entecavir or another standard-of-care HBV nucleos(t)ide analogue for up to 10 years. We assessed long-term outcomes and relationships with virologic response. METHODS: Patients with chronic HBV infection at 299 centers in Asia, Europe, and North and South America were assigned randomly to groups that received entecavir (n = 6216) or an investigator-selected nonentecavir HBV nucleos(t)ide analogue (n = 6162). Study participants were followed up for up to 10 years in hospital-based or community clinics. Key end points were time to adjudicated clinical outcome events and serious adverse events. In a substudy, we examined relationships between these events and virologic response. RESULTS: There were no significant differences between groups in time to event assessments for primary end points including malignant neoplasms, liver-related HBV disease progression, and death. There were no differences between groups in the secondary end points of nonhepatocellular carcinoma malignant neoplasms and hepatocellular carcinoma. In a substudy of 5305 patients in China, virologic response, regardless of treatment group, was associated with a reduced risk of liver-related HBV disease progression (hazard ratio, 0.09; 95% CI, 0.038-0.221) and hepatocellular carcinoma (hazard ratio, 0.03; 95% CI, 0.009-0.113). Twelve patients given entecavir (0.2%) and 50 patients given nonentecavir drugs (0.8%) reported treatment-related serious adverse events. CONCLUSIONS: In a randomized controlled trial of patients with chronic HBV infection, we associated entecavir therapy with a low rate of adverse events over 10 years of follow-up evaluation. Patients receiving entecavir vs another nucleos(t)ide analogue had comparable rates of liver- and non-liver-related clinical outcome events. Participants in a China cohort who maintained a virologic response, regardless of treatment group, had a reduced risk of HBV-related outcome events including hepatocellular carcinoma. ClinicalTrials.gov identifier no: NCT00388674.


Asunto(s)
Hepatitis B Crónica , Neoplasias Hepáticas , Antivirales/efectos adversos , Guanina/análogos & derivados , Virus de la Hepatitis B , Hepatitis B Crónica/tratamiento farmacológico , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/epidemiología , Resultado del Tratamiento
17.
Probiotics Antimicrob Proteins ; 11(4): 1379-1390, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31001786

RESUMEN

Salmonella is an important zoonotic pathogen and is a major cause of gastrointestinal diseases worldwide. The current serious problem of antibiotic abuse has prompted the search for new substitutes for antibiotics. JH-3 is a small antimicrobial peptide with broad-spectrum bactericidal activity. In this study, we showed that JH-3 has good bactericidal activity towards the clinical isolate Salmonella enterica serovar Typhimurium strain CVCC541. The minimum inhibitory concentration (MIC) of JH-3 against this bacterium was determined to be 100 µg/mL, which could decrease the number of CVCC541 cells by 1000-fold in vitro within 5 h. The transmission electron microscopy (TEM) results showed that JH-3 can damage the cell wall and membrane of CVCC541, leading to the leakage of cell contents and subsequent cell death. To measure the bactericidal activity of CVCC541-infected mice were treated intraperitoneally 40 or 10 mg/kg JH-3 at 2 h or 3 days postinfection. Our results showed that treatment with 40 mg/kg JH-3 at 2 h postinfection had the best therapeutic effect and could significantly protect mice from a lethal dose of CVCC541. Furthermore, the clinical symptoms, bacterial burden in blood and organs, and intestinal pathological changes were all decreased and were close to normal. This study examined the therapeutic effect of the antimicrobial peptide JH-3 against S. enterica CVCC541 infection for the first time and determined the therapeutic effect of different JH-3 doses and treatment times, laying the foundation for studies of new antimicrobial agents.


Asunto(s)
Antibacterianos/administración & dosificación , Péptidos/administración & dosificación , Infecciones por Salmonella/tratamiento farmacológico , Salmonella typhimurium/efectos de los fármacos , Animales , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Infecciones por Salmonella/microbiología , Salmonella typhimurium/crecimiento & desarrollo , Salmonella typhimurium/patogenicidad , Virulencia/efectos de los fármacos
18.
Fish Shellfish Immunol ; 88: 53-64, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30790659

RESUMEN

A 58-day feeding trial was conducted to evaluate the effects of dietary myo-inositol (MI) supplementation on growth performance, haematological parameters, hepatopancreas histopathology and antioxidant status of Litopenaeus vannamei fed with oxidized fish oil (OFO). Control diet contained fresh fish oil (FFO) without MI supplementation. The other four diets contained two oxidation levels of OFO (peroxide value: 133.2 and 268.7 meq kg-1) with or without 200 mg MI kg-1 diets (MI0+L, MI0+H, MI200 + L and MI200 + H). Results showed that OFO-supplemented groups (without MI supplementation) showed better growth performance and lower whole-body inositol content when opposed to control group. MI supplementation significantly improved whole-body inositol content in high-oxidized fish oil (HOFO) groups, and also reduced whole-body lipid in low-oxidized fish oil (LOFO) groups. Moreover, Supplementation of OFO and MI markedly hit the fatty acid profile of muscle. HOFO caused severe histopathological changes in hepatopancreas of shrimp, which slightly alleviated by MI supplementation. MI supplementation also grew the total protein (TP) content and alkaline phosphatase (AKP) activity and decreased the activities of aspartate aminotransferase (AST) and alanine aminotransferase (ALT) of serum in OFO-supplemented groups. Ingestion of OFO increased levels of lipid peroxidation and protein oxidation in serum or hepatopancreas, which partly ameliorated by MI supplementation. Activities of antioxidant enzymes exhibited different expression patterns because of OFO and MI. In addition, HOFO markedly increased mRNA expression levels of antioxidant genes including ferritin (FT), thioredoxin (Trx), GPX, glutathione S-transferase (GST) and catalase (CAT) and decreased peroxiredoxin (Prx) expression, in which expression of GPX and Prx were increased owing to MI supplementation. Therefore, it suggested that dietary OFO stimulated growth performance, but also induced oxidative stress and caused impairment to hepatopancreas in L. vannamei. The negative impact brought about by OFO was partially mitigated by dietary MI supplementation.


Asunto(s)
Alimentación Animal/análisis , Aceites de Pescado , Inositol/farmacología , Penaeidae/efectos de los fármacos , Animales , Antioxidantes/análisis , Acuicultura/métodos , Dieta/veterinaria , Hepatopáncreas/efectos de los fármacos , Hepatopáncreas/patología , Peroxidación de Lípido , Oxidación-Reducción , Penaeidae/crecimiento & desarrollo , Penaeidae/metabolismo
19.
Molecules ; 24(3)2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30736473

RESUMEN

The antibiotic resistance of Salmonella has become increasingly serious due to the increased use of antibiotics, and antimicrobial peptides have been considered as an ideal antibiotic alternative. Salmonella can induce macrophage apoptosis and thus further damage the immune system. The antimicrobial peptide JH-3 has been shown to have a satisfactory anti-Salmonella effect in previous research, but its mechanism of action remains unknown. In this study, the effects of JH-3 on macrophages infected with Salmonella Typhimurium CVCC541 were evaluated at the cellular level. The results showed that JH-3 significantly alleviated the damage to macrophages caused by S. Typhi infection, reduced the release of lactic dehydrogenase (LDH), and killed the bacteria in macrophages. In addition, JH-3 decreased the phosphorylation level of p65 and the expression and secretion of interleukin 2 (IL-2), IL-6, and tumor necrosis factor-α (TNF-α) by inhibiting the activation of the mitogen-activated protein kinase (MAPK) (p38) signaling pathway and alleviating the cellular inflammatory response. From confocal laser scanning microscopy and flow cytometry assays, JH-3 was observed to inhibit the release of cytochrome c in the cytoplasm; the expression of TNF-αR2, caspase-9, and caspase-8; to further weaken caspase-3 activation; and to reduce the S.-Typhi-induced apoptosis of macrophages. In summary, the mechanism by which JH-3 inhibits Salmonella infection was systematically explored at the cellular level, laying the foundation for the development and utilization of JH-3 as a therapeutic alternative to antibiotics.


Asunto(s)
Antiinfecciosos/farmacología , Apoptosis/efectos de los fármacos , Citocinas/metabolismo , Mediadores de Inflamación/metabolismo , Péptidos/farmacología , Salmonella typhimurium/efectos de los fármacos , Animales , Antiinfecciosos/química , Biomarcadores , Citocinas/genética , Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Péptidos/química , Células RAW 264.7 , Infecciones por Salmonella/genética , Infecciones por Salmonella/metabolismo , Infecciones por Salmonella/microbiología , Transducción de Señal/efectos de los fármacos , Factor de Transcripción ReIA/metabolismo
20.
Pathol Res Pract ; 215(3): 586-593, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30683475

RESUMEN

Interferon-ß (IFN-ß) exhibits a tumor-killing effect; however, injection of IFN-ß alone for lung cancer is often accompanied by side effects. This study investigated the possibility of using umbilical cord mesenchymal stem cells (MSCs) as cellular carriers of IFN-ß. Isolated umbilical cord MSCs were transfected with a lentivirus packaging IFN-ß-overexpression plasmid. A549 cells were subcutaneously injected into nude mice to establish a non-small cell lung cancer (NSCLC) mouse model. A total of 50 mice were randomly assigned to 5 different groups: a control group, IFN-ß group, IFN-ß-MSCs group, MSCs-lentivirus group, and MSCs group. Next, the IFN-ß-MSCs, MSCs-lentivirus, and MSCs were injected into the A549 lung cancer-bearing mice in the IFN-ß-MSCs, MSCs-lentivirus and MSCs groups, respectively. Mice in the control and IFN-ß groups were injected with solvent or IFN-ß solution. The tumors in nude mice in the IFN-ß and IFN-ß-MSCs groups grew at significantly slower rates than tumors in the control group, and tumors in the MSCs-lentivirus and MSC groups also grew slowly. The rates of tumor cell apoptosis in the IFN-ß and IFN-ß-MSCs groups were significantly higher than those in the MSCs-lentivirus and MSCs groups. The livers, lungs, and kidneys of nude mice in the IFN-ß group displayed hyperemia, exudation, and pathological lesions, while those of nude mice in the IFN-ß-MSCs group showed no abnormal changes. Both INF-ß-MSCs and INF-ß inhibited the growth of subcutaneously implanted lung tumors; however, INF-ß-MSCs specifically targeted the tumor cells, and did not produce the damage to internal organs caused by the use of INF-ß alone.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Terapia Genética/métodos , Interferón beta/administración & dosificación , Neoplasias Pulmonares , Trasplante de Células Madre Mesenquimatosas/métodos , Células A549 , Animales , Vectores Genéticos , Humanos , Interferón beta/genética , Lentivirus , Células Madre Mesenquimatosas , Ratones , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA