Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Mol Biol Rep ; 51(1): 561, 2024 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-38643442

RESUMEN

BACKGROUND: Lysine [K] methyltransferase 2A (KMT2A, previously known as MLL) gene rearrangements are common in acute leukemias of various lineages and are associated with features such as chemotherapy resistance and rapid relapse. KMT2A::CBL is a rare fusion of unknown pathogenesis generated by a unique interstitial deletion of chromosome 11 that has been reported across a wide age range in both acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) patients. The leukemogenic effect of the KMT2A::CBL rearrangement and its association with clinical prognosis have not been well clarified. METHODS AND RESULTS: We report the case of a 64-year-old female who was diagnosed with acute monoblastic leukemia (M5a) and who acquired the rare KMT2A::CBL fusion. The patient received multiple cycles of therapy but did not achieve remission and eventually succumbed to severe infection and disease progression. Additionally, we characterized the predicted KMT2A-CBL protein structure in this case to reveal the underlying leukemogenic mechanisms and summarized reported cases of hematological malignancies with KMT2A::CBL fusion to investigate the correlation of gene rearrangements with clinical outcomes. CONCLUSIONS: This report provides novel insights into the leukemogenic potential of the KMT2A::CBL rearrangement and the correlation between gene rearrangements and clinical outcomes.


Asunto(s)
N-Metiltransferasa de Histona-Lisina , Leucemia Monocítica Aguda , Proteína de la Leucemia Mieloide-Linfoide , Proteínas Proto-Oncogénicas c-cbl , Femenino , Humanos , Persona de Mediana Edad , Progresión de la Enfermedad , Reordenamiento Génico/genética , Leucemia Monocítica Aguda/genética , Leucemia Monocítica Aguda/patología , N-Metiltransferasa de Histona-Lisina/genética , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas Proto-Oncogénicas c-cbl/genética
2.
Cardiovasc Toxicol ; 24(4): 408-421, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38411850

RESUMEN

Halloysite nanotubes (HNTs) are nanomaterials (NMs) derived from natural clays and have been considered as biocompatible NMs for biomedical uses. However, the cardiovascular toxicity of HNTs has not been thoroughly investigated. In this study, we compared the cardiotoxicity of HNTs and multi-walled carbon nanotubes (MWCNTs), focusing on the changes in Kruppel-like factor (KLF)-mediated signaling pathways. Mice were intravenously injected with 50 µg NMs, once a day, for 5 days, and then mouse hearts were removed for experiments. While HNTs or MWCNTs did not induce obvious pathological changes, RNA-sequencing data suggested the alterations of KLF gene expression. We further confirmed an increase of Klf15 positive cells, accompanied by changes in Klf15-related gene ontology (GO) terms. We noticed that most of the changed GO terms are related with the regulation of gene expression, and we confirmed that the NMs increased myoneurin (Mynn) but decreased snail family transcriptional repressor 1 (Snai1), two transcription factors (TFs) related with Klf15. Besides, the changed GO terms also include metal ion binding and positive regulation of glucose import, and we verified an increase of phosphoenolpyruvate carboxykinase 1 (Pck1) and insulin receptor (Insr). However, HNTs and MWCNTs only showed minimal impact on cell death signaling pathways, and no increase in apoptotic sites was observed after NM treatment. We concluded that intravenous administration of HNTs and MWCNTs activated a protective TF, namely Klf15 in mouse aortas, to alter gene expression and signaling pathways related with metal ion binding and glucose import.


Asunto(s)
Nanotubos de Carbono , Animales , Ratones , Nanotubos de Carbono/toxicidad , Arcilla , Inyecciones Intravenosas , Factores de Transcripción de Tipo Kruppel/genética , Glucosa
3.
Toxins (Basel) ; 15(9)2023 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-37756005

RESUMEN

Microcystin-LR (MC-LR) contamination is a worldwide environmental problem that poses a grave threat to the water ecosystem and public health. Exposure to MC-LR has been associated with the development of intestinal injury, but there are no effective treatments for MC-LR-induced intestinal disease. Probiotics are "live microorganisms that are beneficial to the health of the host when administered in sufficient quantities". It has been demonstrated that probiotics can prevent or treat a variety of human diseases; however, their ability to mitigate MC-LR-induced intestinal harm has not yet been investigated. The objective of this study was to determine whether probiotics can mitigate MC-LR-induced intestinal toxicity and its underlying mechanisms. We first evaluated the pathological changes in colorectal tissues using an animal model with sub-chronic exposure to low-dose MC-LR, HE staining to assess colorectal histopathologic changes, qPCR to detect the expression levels of inflammatory factors in colorectal tissues, and WB to detect the alterations on CSF1R signaling pathway proteins in colorectal tissues. Microbial sequencing analysis and screening of fecal microorganisms differential to MC-LR treatment in mice. To investigate the role of microorganisms in MC-LR-induced colorectal injury, an in vitro model of MC-LR co-treatment with microorganisms was developed. Our findings demonstrated that MC-LR treatment induced an inflammatory response in mouse colorectal tissues, promoted the expression of inflammatory factors, activated the CSF1R signaling pathway, and significantly decreased the abundance of Lactobacillus. In a model of co-treatment with MC-LR and Lactobacillus fermentum (L. fermentum), it was discovered that L. fermentum substantially reduced the incidence of the colorectal inflammatory response induced by MC-LR and inhibited the protein expression of the CSF1R signaling pathway. This is the first study to suggest that L. fermentum inhibits the CSF1R signaling pathway to reduce the incidence of MC-LR-induced colorectal inflammation. This research may provide an excellent experimental foundation for the development of strategies for the prevention and treatment of intestinal diseases in MC-LR.


Asunto(s)
Neoplasias Colorrectales , Limosilactobacillus fermentum , Humanos , Animales , Ratones , Ecosistema , Inflamación/inducido químicamente
4.
Chemosphere ; 338: 139455, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37429383

RESUMEN

BACKGROUND: Heavy metals entering the human body could cause damage to a variety of organs. However, the combined harmful effects of exposure to various metals on liver function are not well understood. The purpose of the study was to investigate the independent and joint relationships between heavy metal exposure and liver function in adults. METHODS: The study involved 3589 adults from the National Health and Nutrition Examination Survey. Concentrations of urinary metals, including arsenic (As), cadmium (Cd), lead (Pb), antimony (Sb), barium (Ba), thallium (Tl), tungsten (W), uranium (U), were determined in urine using inductively coupled plasma mass spectrometry. Data for liver function biomarkers included alanine aminotransferase (ALT), aspartate aminotransferase (AST), gamma-glutamyl transaminase (GGT), and alkaline phosphatase (ALP). Survey-weighted linear regression and quantile g-computation (qgcomp) were employed to evaluate the relationship of urinary metals with the markers of liver injury. RESULTS: Cd, U and Ba were found to have positive correlations with ALT, AST, GGT, and ALP in the survey-weighted linear regression analyses. According to the qgcomp analyses, the total metal mixture was positively correlated with ALT (percent change: 8.15; 95% CI: 3.84, 12.64), AST (percent change: 5.55; 95% CI: 2.39, 8.82), GGT (percent change: 14.30; 95% CI: 7.81, 21.18), and ALP (percent change: 5.59; 95% CI: 2.65, 8.62), and Cd, U, and Ba were the main contributors to the combined effects. Positive joint effects were observed between Cd and U on ALT, AST, GGT and ALP, and U and Ba had positive joint effects on ALT, AST and GGT. CONCLUSION: Exposures to Cd, U, and Ba were individually associated with multiple markers of liver injury. Mixed metal exposure might be adversely correlated with markers of liver function. The findings indicated the potential harmful effect of metal exposure on liver function.


Asunto(s)
Cadmio , Metales Pesados , Adulto , Humanos , Encuestas Nutricionales , gamma-Glutamiltransferasa , Hígado , Biomarcadores , Fosfatasa Alcalina
5.
J Appl Toxicol ; 43(11): 1748-1760, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37408164

RESUMEN

Lysine-specific demethylase 1 (LSD1) inhibitors are being developed for cancer therapy, but their bioeffects on vasculatures are not clear. In this study, we compared the influences of ORY-1001 (an LSD1 inhibitor being advanced into clinical trials) and 199 (a novel LSD1 inhibitor recently developed by us) to human umbilical vein endothelial cells (HUVECs) in vitro and further verified the bioeffects of ORY-1001 to zebrafish (Danio rerio) larvae in vivo. The results showed that up to 10 µM ORY-1001 or 199 did not significantly affect the cellular viability of HUVECs but substantially reduced the release of inflammatory interleukin-8 (IL-8) and IL-6. The signaling molecule in vasculatures, NO, was also increased in HUVECs. As the mechanism, the protein levels of endothelial NO synthase (eNOS) or p-eNOS, and their regulators Kruppel-like factor 2 (KLF2) or KLF4, were also increased after drug treatment. In vivo, 24 h treatment with up to 100 nM ORY-1001 reduced blood speed without changing morphologies or locomotor activities in zebrafish larvae. ORY-1001 treatment reduced the expression of il8 but promoted the expression of klf2a and nos in the zebrafish model. These data show that LSD1 inhibitors were not toxic but capable to inhibit inflammatory responses and affect the function of blood vessels through the up-regulation of the NOS-KLF pathway.

6.
Toxins (Basel) ; 15(6)2023 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-37368704

RESUMEN

Previous studies have reported that microcystin-LR (MC-LR) levels are highly correlated with abnormal renal function indicators, suggesting that MC-LR is an independent risk factor for kidney damage. However, the evidence for the exact regulation mechanism of MC-LR on kidney damage is still limited, and further in-depth exploration is needed. In addition, the mitochondria-related mechanism of MC-LR leading to kidney damage has not been elucidated. To this end, the present study aimed to further explore the mechanism of mitophagy related to kidney damage induced by MC-LR through in vitro and in vivo experiments. Male C57BL/6 mice were fed with a standard rodent pellet and exposed daily to MC-LR (20 µg/kg·bw) via intraperitoneal injections for 7 days. Moreover, HEK 293 cells were treated with MC-LR (20 µM) for 24 h. The histopathological results exhibited kidney damage after MC-LR exposure, characterized by structurally damaged nephrotomies, with inflammatory cell infiltration. Similarly, a significant increase in renal interstitial fibrosis was observed in the kidneys of MC-LR-treated mice compared with those of the control group (CT) mice. MC-LR exposure caused impaired kidney function, with markedly increased blood urea nitrogen (BUN), creatinine (Cr), and uric acid (UA) levels in mice. Ultrastructural analysis exhibited obviously swollen, broken, and disappearing mitochondrial crests, and partial mitochondrial vacuoles in the MC-LR-treated HEK 293 cells. The Western blotting results demonstrated that exposure to MC-LR significantly increased the protein expressions of MKK6, p-p38, and p62, while the expression of mitophagy-related proteins was significantly inhibited in the kidneys of mice and HEK293 cells, including parkin, TOM20, and LC3-II, indicating the inhibition of mitophagy. Therefore, our data suggest that the inhibition of MKK6-mediated mitophagy might be the toxicological mechanism of kidney toxicity in mice with acute exposure to MC-LR.


Asunto(s)
Enfermedades Renales , Mitofagia , Animales , Ratones , Masculino , Humanos , Células HEK293 , Ratones Endogámicos C57BL , Riñón , Microcistinas/toxicidad , Enfermedades Renales/inducido químicamente
7.
J Immunother Cancer ; 11(2)2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36808074

RESUMEN

BACKGROUND: Murine chimeric antigen receptor T (CAR-T) cell therapy has demonstrated clinical benefit in patients with relapsed/refractory (R/R) B-cell acute lymphoblastic leukemia (B-ALL). However, the potential immunogenicity of the murine single-chain variable fragment domain may limit the persistence of CAR-T cell, leading to relapse. METHODS: We performed a clinical trial to determine the safety and efficacy of autologous and allogeneic humanized CD19-targeted CAR-T cell (hCART19) for R/R B-ALL. Fifty-eight patients (aged 13-74 years) were enrolled and treated between February 2020 and March 2022. The endpoints were complete remission (CR) rate, overall survival (OS), event-free survival (EFS), and safety. RESULTS: Overall, 93.1% (54/58) of patients achieved CR or CR with incomplete count recovery (CRi) by day 28, with 53 patients having minimal residual disease negativity. With a median follow-up of 13.5 months, the estimated 1-year OS and EFS were 73.6% (95% CI 62.1% to 87.4%) and 46.0% (95% CI 33.7% to 62.8%), with a median OS and EFS of 21.5 months and 9.5 months, respectively. No significant increase in human antimouse antibodies was observed following infusion (p=0.78). Duration of B-cell aplasia in the blood was observed for as long as 616 days, which was longer than that in our prior mCART19 trial. All toxicities were reversible, including severe cytokine release syndrome, which developed in 36% (21/58) of patients and severe neurotoxicity, which developed in 5% (3/58) of patients. Compared with our prior mCART19 trial, patients treated with hCART19 had longer EFS without increased toxicity. Additionally, our data also suggest that patients treated with consolidation therapy, including allogeneic hematopoietic stem cell transplantation or CD22-targeted CAR-T cell, following hCART19 therapy had a longer EFS than those without consolidation therapy. CONCLUSION: hCART19 has good short-term efficacy and manageable toxicity in R/R B-ALL patients. TRIAL REGISTRATION NUMBER: NCT04532268.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Linfoma de Células B , Leucemia-Linfoma Linfoblástico de Células Precursoras , Receptores Quiméricos de Antígenos , Humanos , Animales , Ratones , Receptores Quiméricos de Antígenos/uso terapéutico , Inmunoterapia Adoptiva/efectos adversos , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Supervivencia sin Progresión , Linfoma de Células B/tratamiento farmacológico , Antígenos CD19 , Enfermedad Aguda , Proteínas Adaptadoras Transductoras de Señales
8.
Mol Omics ; 19(2): 150-161, 2023 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-36538054

RESUMEN

Although titanium (Ti)-based nanomaterials (NMs) were traditionally considered as biologically inert materials, it was recently reported that Ti-based NMs induce adverse vascular effects by inhibiting Kruppel-like factor 2 (KLF2) and/or KLF4, vasoprotective KLFs with well-documented regulatory activity in NO signaling. However, the potential roles of other KLFs are not clear. KLF6 was recently identified as an important KLF involved in regulating endothelial dysfunction, inflammation, and angiogenesis, therefore, this study investigated the influence of titanate nanofibers (TiNFs) on KLF6-mediated events. Ingenuity pathway analysis (IPA) showed that TiNFs altered the expression of a panel of KLF6-related genes: KLF6-mediated gene ontology (GO) terms were altered, categories including cytokine-mediated signaling pathways, transcription factor (TF) functions and membrane-bound organelles. Additionally, RT-PCR confirmed that TiNFs increased KLF6 activating transcription factor 3 (ATF3), a TF involved in endoplasmic reticulum (ER) stress, and ELISA confirmed the increase of soluble monocyte chemotactic protein 1 (sMCP-1), a KLF6-related inflammatory cytokine. Interestingly, the activation of klf6, atf3 and C-C motif chemokine ligand 2 (ccl2; mcp-1 encoding gene) was observed in aortas of mice following one-time intravenous injection but not intratracheal instillation of TiNFs (100 µg per mouse), indicating a need for direct contact with NMs to activate klf6-mediated pathways in vivo. In endothelial cells, KLF6 knockdown inhibited the expression of ATF3 but not CCL2, suggesting the regulatory role of KLF6 in ATF3 expression. Overall, this study uncovered a previously unknown role of KLF6 in TiNF-induced vascular effects both in vitro and in vivo.


Asunto(s)
Factor de Transcripción Activador 3 , Nanofibras , Ratones , Animales , Factor de Transcripción Activador 3/genética , Factor de Transcripción Activador 3/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Células Endoteliales , Aorta/metabolismo , Citocinas
9.
J Appl Toxicol ; 42(11): 1822-1831, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35727742

RESUMEN

Recent studies revealed a causal relationship between Toll-like receptors (TLRs) and lipid droplet biogenesis. Interestingly, it has been reported before that nanomaterials (NMs) were capable to modulate TLRs, but it remains unclear if NMs could affect lipid levels via TLR signaling pathways. In this study, we investigated the influences of airway exposure to graphene oxide (GO) on TLR3 signaling pathways and lipid levels in mouse livers. Intratracheal instillation of GO (0.1, 1, and 5 mg/kg, once a day, totally 5 days) induced inflammatory cell infiltrations as indicated by hematoxylin-eosin (H&E) staining and fibrosis as indicated by Masson staining in lungs, accompanying with decreased TLR3 proteins. Consistently, a TLR3-regulated anti-virus protein, namely interferon induced protein with tetratricopeptide repeats 1 (IFIT1), as well as two TLR3-regulated lipid proteins, namely radical S-adenosyl methionine domain containing 2 (RSAD2) and perilipin 2 (PLIN2), were decreased in lungs. The protein levels of interferon-ß in serum were also decreased. In livers, GO exposure induced disorganization of liver cells but not fibrosis. In agreement with the trends observed in lungs, TLR3, IFIT1, RSAD2, and PLIN2 proteins were decreased in livers. As a possible consequence, GO exposure dose-dependently decreased lipid levels in livers as indicated by oil red O and BODIPY 493/503 staining. We concluded that airway exposure to GO decreased anti-virus responses and lipid levels in mouse livers via the suppression of TLR3.


Asunto(s)
Receptor Toll-Like 3 , Receptores Toll-Like , Animales , Eosina Amarillenta-(YS) , Grafito , Hematoxilina , Interferón beta/metabolismo , Interferones/metabolismo , Lípidos , Hígado/metabolismo , Metionina , Ratones , Perilipina-2 , Receptor Toll-Like 3/metabolismo , Receptores Toll-Like/metabolismo
10.
Virulence ; 13(1): 241-257, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35067196

RESUMEN

Numerous viruses manipulate host factors for viral production. We demonstrated that human enterovirus A71 (EVA71), a primary causative agent for hand, foot, and mouth disease (HFMD), increased the level of the DNA damage response (DDR) marker γ-H2AX. DDR is primarily mediated by the ataxia telangiectasia mutated (ATM), ATM and Rad3-related (ATR), or DNA-dependent protein kinase (DNA-PK) pathways. Upregulation of γ-H2AX by EVA71 was dependent on the ATR but not the ATM or DNA-PK pathway. As a nuclear factor, there is no previous evidence of cytoplasmic distribution of γ-H2AX. However, the present findings demonstrated that EVA71 encouraged the localization of γ-H2AX to the cytoplasm. Of note, γ-H2AX formed a complex with structural protein VP3, non-structural protein 3D, and the viral genome. Treatment with an inhibitor or CRISPR/Cas9 technology to decrease or silence the expression of γ-H2AX decreased viral genome replication in host cells; this effect was accompanied by decreased viral protein expression and virions. In animal experiments, caffeine was used to inhibit DDR; the results revealed that caffeine protected neonatal mice from death after infection with EVA71, laying the foundation for new therapeutic applications of caffeine. More importantly, in children with HFMD, γ-H2AX was upregulated in peripheral blood lymphocytes. The consistent in vitro and in vivo data on γ-H2AX from this study suggested that caffeine or other inhibitors of DDR might be novel therapeutic agents for HFMD.


Asunto(s)
Infecciones por Enterovirus , Enterovirus , Histonas , Animales , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Cafeína , ADN , Daño del ADN , Enterovirus/fisiología , Infecciones por Enterovirus/genética , Infecciones por Enterovirus/metabolismo , Histonas/genética , Histonas/metabolismo , Interacciones Microbiota-Huesped , Ratones , Proteínas Virales/genética , Replicación Viral
11.
J Microbiol ; 58(9): 812-820, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32870487

RESUMEN

Enterovirus D68 (EVD68) is an emerging pathogen that recently caused a large worldwide outbreak of severe respiratory disease in children. However, the relationship between EVD68 and host cells remains unclear. Caspases are involved in cell death, immune response, and even viral production. We found that caspase-3 was activated during EVD68 replication to induce apoptosis. Caspase-3 inhibitor (Z-DEVD-FMK) inhibited viral production, protected host cells from the cytopathic effects of EVD68 infection, and prevented EVD68 from regulating the host cell cycle at G0/G1. Meanwhile, caspase-3 activator (PAC-1) increased EVD68 production. EVD68 infection therefore activates caspase-3 for virus production. This knowledge provides a potential direction for the prevention and treatment of disease related to EVD68.


Asunto(s)
Antivirales/farmacología , Caspasa 3/efectos de los fármacos , Caspasa 3/metabolismo , Enterovirus Humano D/efectos de los fármacos , Enterovirus Humano D/crecimiento & desarrollo , Apoptosis/fisiología , Línea Celular Tumoral , Infecciones por Enterovirus/patología , Infecciones por Enterovirus/prevención & control , Infecciones por Enterovirus/virología , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Humanos , Hidrazonas/farmacología , Oligopéptidos/farmacología , Piperazinas/farmacología
12.
Phytother Res ; 33(1): 174-186, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30346067

RESUMEN

Human enterovirus 68 (EVD68) is a primary causative agent for respiratory illness worldwide. Until now, there has been no available medication for treating EVD68-related diseases. Rheum emodin, artemisinin, astragaloside, pseudolaric acid B, oridonin, and erianin are natural extracts from Chinese herbs that have traditionally been used for the treatment and prevention of epidemic diseases. Our results showed that pseudolaric acid B protected cells from EVD68-induced cytopathic effects and decreased viral production. However, the same effects were not observed with rheum emodin, astragaloside, or artemisinin. Pseudolaric acid B inhibited EVD68 production by manipulating the host cell cycle in G2/M phase. Further, either oridonin or erianin related G2/M arrest also inhibited viral production. Due to inducing G2/M phase arrest, pseudolaric acid B, oridonin, and erianin might be good candidates for inhibiting EVD68 production, and Chinese herbs with natural compounds inducing G2/M arrest should be considered for the treatment of EVD68-related diseases.


Asunto(s)
Medicamentos Herbarios Chinos/uso terapéutico , Enterovirus Humano D/patogenicidad , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Humanos
13.
Artículo en Inglés | MEDLINE | ID: mdl-30159255

RESUMEN

Recent epidemiological data indicate that outbreaks of hand, foot, and mouth disease (HFMD), which can be categorized according to its clinical symptoms as typical or atypical, have markedly increased worldwide. A primary causative agent for typical HFMD outbreaks, enterovirus 71 (EV71), has been shown to manipulate the cell cycle in S phase for own replication; however, it is not clear whether coxsackievirus (CVA6), the main agent for atypical HFMD, also regulates the host cell cycle. In this study, we demonstrate for the first time that CVA6 infection arrests the host cell cycle in G0/G1-phase. Furthermore, synchronization in G0/G1 phase, but not S phase or G2/M phase, promotes viral production. To investigate the mechanism of cell cycle arrest induced by CVA6 infection, we analyzed cell cycle progression after cell cycle synchronization at G0/G1 or G2/M. Our results demonstrate that CVA6 infection promotes G0/G1 phase entry from G2/M phase, and inhibits G0/G1 exit into S phase. In line with its role to arrest cells in G0/G1 phase, the expression of cyclinD1, CDK4, cyclinE1, CDK2, cyclinB1, CDK1, P53, P21, and P16 is regulated by CVA6. Finally, the non-structural proteins of CVA6, RNA-dependent RNA polymerase 3D and protease 3C , are demonstrated to be responsible for the G0/G1-phase arrest. These findings suggest that CVA6 infection arrested cell cycle in G0/G1-phase via non-structural proteins 3D and 3C, which may provide favorable environments for virus production.


Asunto(s)
Puntos de Control del Ciclo Celular , Enterovirus/crecimiento & desarrollo , Fase G1 , Interacciones Huésped-Patógeno , Replicación Viral , Línea Celular , Humanos , Proteínas Virales/metabolismo
14.
Front Microbiol ; 9: 817, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29755438

RESUMEN

Previous studies demonstrate that human enterovirus 71 (EV71), a primary causative agent for hand, foot, and mouth disease, activates caspase-3 through the non-structural viral 3C protein to induce host cell apoptosis; however, until now it was unclear how 3C activates caspase-3 and how caspase-3 activation affects viral production. Our results demonstrate that 3C binds caspase-8 and caspase-9 but does not directly bind caspase-3 to activate them, and that the proteolytic activity of 3C is required by the activation of caspase-8, caspase-9, and caspase-3. Inhibition of caspase-3 activity attenuates apoptosis in 3C-transfected cells. Furthermore, caspase-3 inhibitor protects host cells from the cytopathic effect of EV71 infection and prevents cell cycle arrest, which is known to be favored for EV71 viral replication. Inhibition of caspase-3 activity decreases EV71 viral protein expression and viral production, but has no effect on viral entry, replication, even polyprotein translation. Therefore, caspase-3 is exploited functionally by EV71 to facilitate its production, which suggests a novel therapeutic approach for the treatment and prevention of hand, foot, and mouth disease.

15.
Oncol Lett ; 15(1): 863-870, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29399151

RESUMEN

Autophagy and apoptosis are closely associated. In previous studies, pseudolaric acid B (PAB), a diterpene acid isolated from the root and trunk bark of Pseudolarix kaempferi Gordon (Pinaceae), was demonstrated to induce apoptosis in various cell lines. However, in L929 murine fibrosarcoma and SW579 human thyroid squamous cell carcinoma cells, only autophagy was induced. In the present study, another cell line, MRC5 human lung fibroblast cells, was identified in which PAB only induced autophagy. The relationship between apoptosis and autophagy subsequent to PAB treatment in MRC5 cells was explored. When autophagy was inhibited by 3-methyladenine (3MA), apoptosis was induced in the PAB-treated MRC5 cells. To study the mechanism for the promotion of apoptosis by 3MA in the PAB-treated cells, the expression of members from the apoptotic signal pathways was assessed. As Bcl-2, Bcl-2 associated X and pro-caspase-9 expression following PAB treatment was not affected by 3MA treatment, it was determined that apoptosis was induced independent of the mitochondrial pathway of apoptosis. As Fas and pro-caspase-8 expression following PAB treatment were not altered by 3MA, it was further determined that the death receptor pathway was not induced. However, the phosphorylation of c-Jun-N-terminal kinase and the expression of pro-caspase-3 were upregulated, and the phosphorylation of extracellular signal-regulated kinase downregulated, by the combination of PAB and 3MA treatment compared with PAB alone. It was also observed that 3MA did not affect the microtubule aggregation ability of PAB. Therefore, inhibiting autophagy in MRC5 cells did not affect the role of PAB in microtubule aggregation, while apoptosis was induced. This may present a strategy to enhance the anti-tumor effects of PAB.

16.
Oncol Rep ; 38(4): 2211-2218, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28765951

RESUMEN

Human neuroglioma is one of the most common malignant intracranial tumors in neurosurgery, and accounts for more than 50% of all brain cancer cases. Thus, a clinically effective drug with which to treat neuroglioma is urgently required. Pseudolaric acid B (PAB), a diterpene acid isolated from the root and trunk bark of Pseudolarix kaempferi Gordon (Pinaceae), was found to inhibit cell growth in a variety of cancer cell lines, but to date the effect of PAB on neuroglioma remains unclear. MTT analysis confirmed that PAB inhibited neuroglioma A172 cell growth in a time- and dose-dependent manner. In addition, PAB influenced the aggregation of tubulin in A172 cells. Meanwhile following PAB treatment, a higher percentage of cells accumulated in the G2/M phase from 12 to 48 h, while at 36 h, cell cycle slippage into the G0/G1 phase, and at 48 h, slippage into the S phase was observed using flow cytometric analysis. Corresponding protein expression was consistent with the cell cycle alteration as detected by western blotting, and it was speculated that cell cycle slippage was related to reduced effectiveness of PAB which warrants further investigation. Meanwhile PAB induced cell death by regulating p38, ERK and JNK expression and activating the DNA damage response. Therefore, PAB plays an antitumor role in A172 cells, and may be a candidate drug for neuroglioma therapy.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Daño del ADN/efectos de los fármacos , Diterpenos/administración & dosificación , Glioma/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Proliferación Celular/efectos de los fármacos , Diterpenos/química , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/genética , Glioma/patología , Humanos , MAP Quinasa Quinasa 4/genética , Sistema de Señalización de MAP Quinasas , Pinaceae/química , Extractos Vegetales/química , Proteínas Quinasas p38 Activadas por Mitógenos/genética
17.
Artículo en Inglés | MEDLINE | ID: mdl-28229049

RESUMEN

Enterovirus D68 (EV-D68) is an emerging pathogen that recently caused a large outbreak of severe respiratory disease in the United States and other countries. Little is known about the relationship between EV-D68 virus and host cells. In this study, we assessed the effect of the host cell cycle on EV-D68 viral production, as well as the ability of EV-D68 to manipulate host cell cycle progression. The results suggest that synchronization in G0/G1 phase, but not S phase, promotes viral production, while synchronization in G2/M inhibits viral production. Both an early EV-D68 isolate and currently circulating strains of EV-D68 can manipulate the host cell cycle to arrest cells in the G0/G1 phase, thus providing favorable conditions for virus production. Cell cycle regulation by EV-D68 was associated with corresponding effects on the expression of cyclins and CDKs, which were observed at the level of the protein and/or mRNA. Furthermore, the viral non-structural protein 3D of EV-D68 prevents progression from G0/G1 to S. Interestingly, another member of the Picornaviridae family, EV-A71, differs from EV-D68 in that G0/G1 synchronization inhibits, rather than promotes, EV-A71 viral replication. However, these viruses are similar in that G2/M synchronization inhibits the production and activity of both viruses, which is suggestive of a common therapeutic target for both types of enterovirus. These results further clarify the pathogenic mechanisms of enteroviruses and provide a potential strategy for the treatment and prevention of EV-D68-related disease.


Asunto(s)
Ciclo Celular , Enterovirus Humano D/fisiología , Infecciones por Enterovirus/virología , Replicación Viral , Puntos de Control del Ciclo Celular , Línea Celular , Células Cultivadas , Interacciones Huésped-Patógeno , Humanos
18.
Acta Pharmacol Sin ; 38(3): 392-401, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27840410

RESUMEN

Human enterovirus 71 (EV71) is the primary causative agent of recent large-scale outbreaks of hand, foot, and mouth disease (HFMD) in Asia. Currently, there are no drugs available for the prevention and treatment of HFMD. In this study, we compared the anti-EV71 activities of three natural compounds, rheum emodin, artemisinin and astragaloside extracted from Chinese herbs Chinese rhubarb, Artemisia carvifolia and Astragalus, respectively, which have been traditionally used for the treatment and prevention of epidemic diseases. Human lung fibroblast cell line MRC5 was mock-infected or infected with EV71, and treated with drugs. The cytotoxicity of the drugs was detected with MTT assay. The cytopathic effects such as cell death and condensed nuclei were morphologically observed. The VP1-coding sequence required for EV71 genome replication was assayed with qRT-PCR. Viral protein expression was analyzed with Western blotting. Viral TCID50 was determined to evaluate EV71 virulence. Flow cytometry analysis of propidium iodide staining was performed to analyze the cell cycle distribution of MRC5 cells. Rheum emodin (29.6 µmol/L) effectively protected MRC5 cells from EV71-induced cytopathic effects, which resulted from the inhibiting viral replication: rheum emodin treatment decreased viral genomic levels by 5.34-fold, viral protein expression by less than 30-fold and EV71 virulence by 0.33107-fold. The fact that inhibition of rheum emodin on viral virulence was much stronger than its effects on genomic levels and viral protein expression suggested that rheum emodin inhibited viral maturation. Furthermore, rheum emodin treatment markedly diminished cell cycle arrest at S phase in MRC5 cells, which was induced by EV71 infection and favored the viral replication. In contrast, neither astragaloside (50 µmol/L) nor artemisinin (50 µmol/L) showed similar anti-EV71 activities. Among the three natural compounds tested, rheum emodin effectively suppressed EV71 viral replication, thus is a candidate anti-HFMD drug.


Asunto(s)
Antivirales/farmacología , Ciclo Celular/efectos de los fármacos , Emodina/farmacología , Enterovirus Humano A/efectos de los fármacos , Animales , Artemisininas/farmacología , Chlorocebus aethiops , Enterovirus Humano A/fisiología , Interacciones Huésped-Patógeno , Humanos , Fase S/efectos de los fármacos , Células Vero , Replicación Viral/efectos de los fármacos
19.
Oncol Rep ; 37(1): 519-525, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27878296

RESUMEN

High hepatitis B virus (HBV) load and chronic hepatitis B infection increase the risk of developing hepatocellular carcinoma (HCC), and is also associated with recurrence of HBV-related HCC. The aim of the present study was to investigate whether pseudolaric acid B (PAB), a diterpene acid isolated from the root and trunk bark of Pseudolarix kaempferi Gordon (Pinaceae), has an inhibitory role on the HBV secretion in HBV-related HCC. By detecting HBV surface antigen (HBsAg) by ELISA it was found that PAB inhibited HBV secretion in HepG2215 compared to control group, but did not decrease the intracellular HBV level, and the results were repeated in HepG2 cell transfect with HBV gene. Therefore, our results proved that PAB had the ability to inhibit HBV secretion. Moreover, it was shown that HepG2215 cells with HBV gene accumulated more in G0/G1 phase than HepG2 cells without HBV gene through detecting cell cycle distribution by flow cytometry, which indicated that HBV replication might favor the cell cycle environment of G0/G1 phase. However, HepG2 cells entered G2/M phase earlier than HepG2215 when PAB treatment induced G2/M arrest, therefore, HBV retarded the entry of G2/M to sustain the status of G0/G1 phase, while PAB finally changed the cell cycle environment favored by HBV virus. In addition, PAB also induced HepG2215 cell apoptosis, which would be helpful to kill the cells infected by HBV and help for devouring HBV by macrophage. Therefore, PAB inhibited HBV secretion through apoptosis and cell cycle arrest. The present findings contribute to a future potential chemotherapeutic drug in the treatment of HBV-related HCC.


Asunto(s)
Diterpenos/farmacología , Virus de la Hepatitis B/efectos de los fármacos , Liberación del Virus/efectos de los fármacos , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/prevención & control , Carcinoma Hepatocelular/virología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Hep G2 , Virus de la Hepatitis B/fisiología , Humanos , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/prevención & control , Neoplasias Hepáticas/virología , Replicación Viral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA