Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Control Release ; 370: 501-515, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38703950

RESUMEN

Although nanomaterial-based nanomedicine provides many powerful tools to treat cancer, most focus on the "immunosilent" apoptosis process. In contrast, ferroptosis and immunogenic cell death, two non-apoptotic forms of programmed cell death (PCD), have been shown to enhance or alter the activity of the immune system. Therefore, there is a need to design and develop nanoplatforms that can induce multiple modes of cell death other than apoptosis to stimulate antitumor immunity and remodel the immunosuppressive tumor microenvironment for cancer therapy. In this study, a new type of multifunctional nanocomposite mainly consisting of HMME, Fe3+ and Tannic acid, denoted HFT NPs, was designed and synthesized to induce multiple modes of cell death and prime the tumor microenvironment (TME). The HFT NPs consolidate two functions into one nano-system: HMME as a sonosensitizer for the generation of reactive oxygen species (ROS) 1O2 upon ultrasound irradiation, and Fe3+ as a GSH scavenger for the induction of ferroptosis and the production of ROS ·OH through inorganic catalytic reactions. The administration of HFT NPs and subsequent ultrasound treatment caused cell death through the consumption of GSH, the generation of ROS, ultimately inducing apoptosis, ferroptosis, and immunogenic cell death (ICD). More importantly, the combination of HFT NPs and ultrasound irradiation could reshape the TME and recruit more T cell infiltration, and its combination with immune checkpoint blockade anti-PD-1 antibody could eradicate tumors with low immunogenicity and a cold TME. This new nano-system integrates sonodynamic and chemodynamic properties to achieve outstanding therapeutic outcomes when combined with immunotherapy. Collectively, this study demonstrates that it is possible to potentiate cancer immunotherapy through the rational and innovative design of relatively simple materials.


Asunto(s)
Ferroptosis , Inmunoterapia , Especies Reactivas de Oxígeno , Microambiente Tumoral , Microambiente Tumoral/efectos de los fármacos , Animales , Inmunoterapia/métodos , Especies Reactivas de Oxígeno/metabolismo , Línea Celular Tumoral , Ferroptosis/efectos de los fármacos , Humanos , Muerte Celular/efectos de los fármacos , Apoptosis/efectos de los fármacos , Ratones Endogámicos C57BL , Taninos/química , Taninos/farmacología , Ratones , Femenino , Nanocompuestos/química , Nanocompuestos/administración & dosificación , Ondas Ultrasónicas , Neoplasias/terapia , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Neoplasias/patología , Muerte Celular Inmunogénica/efectos de los fármacos
2.
Mol Cancer ; 22(1): 125, 2023 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-37543582

RESUMEN

Hairy cell leukemia (HCL) is a B-lymphoma induced by BRAF(V600E) mutation. However, introducing BRAF(V600E) in B-lymphocytes fails to induce hematological malignancy, suggesting that BRAF(V600E) needs concurrent mutations to drive HCL ontogeny. To resolve this issue, here we surveyed human HCL genomic sequencing data. Together with previous reports, we speculated that the tumor suppressor TP53, P27, or PTEN restrict the oncogenicity of BRAF(V600E) in B-lymphocytes, and therefore that their loss-of-function facilitates BRAF(V600E)-driven HCL ontogeny. Using genetically modified mouse models, we demonstrate that indeed BRAF(V600E)KI together with Trp53KO or pTENKO in B-lymphocytes induces chronic lymphoma with pathological features of human HCL. To further understand the cellular programs essential for HCL ontogeny, we profiled the gene expression of leukemic cells isolated from BRAF(V600E)KI and Trp53KO or pTENKO mice, and found that they had similar but different gene expression signatures that resemble that of M2 or M1 macrophages. In addition, we examined the expression signature of transcription factors/regulators required for germinal center reaction and memory B cell versus plasma cell differentiation in these leukemic cells and found that most transcription factors/regulators essential for these programs were severely inhibited, illustrating why hairy cells are arrested at a transitional stage between activated B cells and memory B cells. Together, our study has uncovered concurrent mutations required for HCL ontogeny, revealed the B cell origin of hairy cells and investigated the molecular basis underlying the unique pathological features of the disease, with important implications for HCL research and treatment.


Asunto(s)
Leucemia de Células Pilosas , Animales , Humanos , Ratones , Linfocitos B/metabolismo , Leucemia de Células Pilosas/genética , Leucemia de Células Pilosas/metabolismo , Leucemia de Células Pilosas/patología , Mutación , Proteínas Proto-Oncogénicas B-raf , Factores de Transcripción/genética
3.
J Control Release ; 361: 427-442, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37487929

RESUMEN

Due to the unique physicochemical properties, mesoporous silica nanoparticles (MONs) have been widely utilized in biomedical fields for drug delivery, gene therapy, disease diagnosis and imaging. With the extensive applications and large-scale production of MONs, the potential effects of MONs on human health are gaining increased attention. To better understand the cellular and molecular mechanisms underlying the effects of MONs on the mouse liver, we profiled the transcriptome of 63,783 single cells from mouse livers following weekly intravenous administration of MONs for 2 weeks. The results showed that the proportion of endothelial cells and CD4+ T cells was increased, whereas that of Kupffer cells was decreased, in a dose-dependent manner after MONs treatment in the mouse liver. We also observed that the proportion of inflammation-related Kupffer cell subtype and wound healing-related hepatocyte subtype were elevated, but the number of hepatocytes with detoxification characteristics was reduced after MONs treatment. The cell-cell communication network revealed that there was more crosstalk between cholangiocytes and Kupffer cells, liver capsular macrophages, hepatic stellate cells, and endothelial cells following MONs treatment. Furthermore, we identified key ligand-receptor pairs between crucial subtypes after MONs treatment that are known to promote liver fibrosis. Collectively, our study explored the effects of MONs on mouse liver at a single-cell level and provides comprehensive information on the potential hepatotoxicity of MONs.


Asunto(s)
Células Endoteliales , Nanopartículas , Ratones , Humanos , Animales , Dióxido de Silicio/química , Transcriptoma , Hígado , Hepatocitos , Nanopartículas/química
5.
Med Res Rev ; 41(6): 3096-3117, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33599316

RESUMEN

Ever since the discovery of insulin, natural peptides have become an important resource for therapeutic development. Decades of research has led to the discovery of a long list of peptide drugs with broad applications in clinics, from antibiotics to hypertension treatment to pain management. Many of these US FDA-approved peptide drugs are derived from microorganisms and animals. By contrast, the great potential of plant cyclic peptides as therapeutics remains largely unexplored. These macrocyclic peptides typically have rigid structures, good bioavailability and membrane permeability, making them appealing candidates for drug development and engineering. In this review, we introduce the three major classes of plant cyclic peptides and summarize their potential medical applications. We discuss how we can leverage the genome information of many different plants to quickly search for new cyclic peptides and how we can take advantage of the insights gained from their biosynthetic pathways to transform the process of production and drug development. These recent developments have provided a new angle for exploring and exploiting plant cyclic peptides, and we believe that many more peptide drugs derived from plants are about to come.


Asunto(s)
Descubrimiento de Drogas , Péptidos Cíclicos , Plantas/química , Humanos , Péptidos Cíclicos/química , Péptidos Cíclicos/farmacología
6.
Pharmacol Ther ; 216: 107673, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32926934

RESUMEN

Aging is a natural biological progress accompanied by the gradual decline in physiological functions, manifested by its close association with an increased incidence of human diseases and higher vulnerability to death. Those diseases include neurological disorders, cardiovascular diseases, diabetes, and cancer, many of which are currently without effective cures. Even though aging is inevitable, there are still interventions that can be developed to prevent/delay the onset and progression of those aging-associated diseases and extend healthspan and/or lifespan. Here, we review decades of research that reveals the molecular pathways underlying aging and forms the biochemical basis for anti-aging drug development. Importantly, due to the vast chemical space of natural products and the rich history of herb medicines in treating human diseases documented in different cultures, natural products have played essential roles in aging research. Using several of the most promising natural products and their derivatives as examples, we discuss how natural products serve as an inspiration resource that helped the identification of key components/pathways underlying aging, their mechanisms of action inside the cell, and the functional scaffolds or targeting mechanisms that can be learned from natural products for drug engineering and optimization. We argue that natural products might eventually provide a solution to aging and aging-associated diseases.


Asunto(s)
Envejecimiento/efectos de los fármacos , Productos Biológicos/uso terapéutico , Enfermedades Cardiovasculares/tratamiento farmacológico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Preparaciones de Plantas/uso terapéutico , Adolescente , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Productos Biológicos/efectos adversos , Productos Biológicos/aislamiento & purificación , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Niño , Preescolar , Diabetes Mellitus Tipo 2/epidemiología , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Femenino , Humanos , Incidencia , Lactante , Masculino , Persona de Mediana Edad , Neoplasias/epidemiología , Neoplasias/metabolismo , Neoplasias/patología , Enfermedades del Sistema Nervioso/epidemiología , Enfermedades del Sistema Nervioso/metabolismo , Enfermedades del Sistema Nervioso/patología , Preparaciones de Plantas/efectos adversos , Preparaciones de Plantas/aislamiento & purificación , Factores de Riesgo , Transducción de Señal , Adulto Joven
7.
J Hematol Oncol ; 13(1): 113, 2020 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-32807225

RESUMEN

Cancer is characterized as a complex disease caused by coordinated alterations of multiple signaling pathways. The Ras/RAF/MEK/ERK (MAPK) signaling is one of the best-defined pathways in cancer biology, and its hyperactivation is responsible for over 40% human cancer cases. To drive carcinogenesis, this signaling promotes cellular overgrowth by turning on proliferative genes, and simultaneously enables cells to overcome metabolic stress by inhibiting AMPK signaling, a key singular node of cellular metabolism. Recent studies have shown that AMPK signaling can also reversibly regulate hyperactive MAPK signaling in cancer cells by phosphorylating its key components, RAF/KSR family kinases, which affects not only carcinogenesis but also the outcomes of targeted cancer therapies against the MAPK signaling. In this review, we will summarize the current proceedings of how MAPK-AMPK signalings interplay with each other in cancer biology, as well as its implications in clinic cancer treatment with MAPK inhibition and AMPK modulators, and discuss the exploitation of combinatory therapies targeting both MAPK and AMPK as a novel therapeutic intervention.


Asunto(s)
Adenilato Quinasa/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Terapia Molecular Dirigida , Proteínas de Neoplasias/fisiología , Neoplasias/enzimología , Aminoácidos/metabolismo , Antineoplásicos/uso terapéutico , Autofagia , Diferenciación Celular/fisiología , División Celular/fisiología , Ensayos Clínicos como Asunto , Sinergismo Farmacológico , Metabolismo Energético , Activación Enzimática , Homeostasis , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Fosforilación , Inhibidores de Proteínas Quinasas/uso terapéutico , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas p21(ras)/antagonistas & inhibidores , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/fisiología , Proteínas Supresoras de Tumor/fisiología , Quinasas raf/antagonistas & inhibidores , Quinasas raf/genética , Quinasas raf/fisiología
8.
Front Pharmacol ; 11: 178, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32180726

RESUMEN

Lung cancer remains a leading cause of cancer-associated mortality worldwide, however, molecular mechanisms underlying lung cancer tumorigenesis and progression remain unknown. Here, we report evidence showing that one member of the mammalian methyltransferase-like family (METTL), METTL7B, is a potential molecular target for treatment of non-small cell lung cancer (NSCLC). METTL7B expression was elevated in the majority of NSCLC comparing to normal tissues. Increased expression of METTL7B contributed to advanced stages of tumor development and poor survival in NSCLC patients. Lentivirus-mediated shRNA silencing of METTL7B suppressed proliferation and tumorigenesis of cancer cells in vitro and in vivo. Investigation on gene expression profiles of NSCLC cells revealed that abundant cell cycle related genes were downregulated in the absence of METTL7B. Pathway enrichment analysis indicated that METTL7B participated in cell cycle regulation. Notably, CCND1, a key regulator for G1/S transition, was significantly decreased with the depletion of METTL7B, resulting in G0/G1 arrest, indicating that METTL7B is critical for cell cycle progression. Taken together, our findings implicate that METTL7B is essential for NSCLC development and progression. METTL7B might serve as a potential therapeutic target for NSCLC.

9.
Cancer Lett ; 476: 13-22, 2020 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-32044357

RESUMEN

Colorectal cancer (CRC), a common malignancy, is among the leading causes of cancer-related deaths worldwide. Developing novel biomarkers is an important public health strategy to effectively reduce the mortality of this disease. Recent studies have found that exosomes may be important sources of biomarkers in CRC. Exosomes are nanometer-sized membrane vesicles (30-200 nm) secreted by normal or cancer cells, which participate in intercellular communication by transporting RNAs and proteins. Accumulating evidence has shown that some differentially expressed RNAs and proteins in exosomes play key roles in the initiation and development of CRC and are potential candidates for malignancy detection. Accordingly, exploring the correlation between these exosomes and CRC may be beneficial for the development of novel biomarkers in this disease. Here, we summarize the important roles of exosomes as biomarkers in CRC diagnosis, as well as the application in the metastasis, chemoresistance, and recrudescence of CRC. In particular, we discuss the prospects and limitations of exosomes as tumor markers.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Colorrectales/diagnóstico , Resistencia a Antineoplásicos/genética , Exosomas/genética , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Humanos , Pronóstico
10.
J Vis Exp ; (149)2019 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-31380842

RESUMEN

The rapidly accelerated fibrosarcoma (RAF) family kinases play a central role in cell biology and their dysfunction leads to cancers and developmental disorders. A characterization of disease-related RAF mutants will help us select appropriate therapeutic strategies for treating these diseases. Recent studies have shown that RAF family kinases have both catalytic and allosteric activities, which are tightly regulated by dimerization. Here, we constructed a set of practical and feasible methods to determine the catalytic and allosteric activities and the relative dimer affinity/stability of RAF family kinases and their mutants. Firstly, we amended the classical in vitro kinase assay by reducing the detergent concentration in buffers, utilizing a gentle quick wash procedure, and employing a glutathione S-transferase (GST) fusion to prevent RAF dimers from dissociating during purification. This enables us to measure the catalytic activity of constitutively active RAF mutants appropriately. Secondly, we developed a novel RAF co-activation assay to evaluate the allosteric activity of kinase-dead RAF mutants by using N-terminal truncated RAF proteins, eliminating the requirement of active Ras in current protocols and thereby achieving a higher sensitivity. Lastly, we generated a unique complementary split luciferase assay to quantitatively measure the relative dimer affinity/stability of various RAF mutants, which is more reliable and sensitive compared to the traditional co-immunoprecipitation assay. In summary, these methods have the following advantages: (1) user-friendly; (2) able to carry out effectively without advanced equipment; (3) cost-effective; (4) highly sensitive and reproducible.


Asunto(s)
Proteínas Serina-Treonina Quinasas/metabolismo , Quinasas raf/metabolismo , Animales , Humanos , Mutación
11.
Breast Cancer Res Treat ; 177(1): 145-153, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31119570

RESUMEN

PURPOSE: PARP4 has been proposed as a candidate breast cancer susceptibility gene. However, its function and involvement in breast carcinogenesis is unclear. We sought to determine the variant frequency of PARP4 in BRCA-negative women referred for genetic testing from Singapore and to perform functional analyses of PARP4. METHODS: Next-generation sequencing of PARP4 was conducted for 198 BRCA-negative cases from Singapore. Three independent case-control association analyses of PARP4 were performed for (1) our Singaporean cohort, (2) three dbGaP datasets, and (3) cases from TCGA, with controls from the Exome Aggregation Consortium (ExAC). PARP4 knockout cells were generated utilizing the CRISPR-Cas9 approach in MDA-MB-231 (breast cancer) and MCF10A (normal breast) cell lines, and colony formation, cell proliferation, and migration assays carried out. RESULTS: Candidate variants in PARP4 were identified in 5.5% (11/198) of our Singapore cohort. Case-control association studies for our cases and the dbGaP datasets showed no significant association. However, a significant association was observed for PARP4 variants when comparing 988 breast cancer cases from the TCGA provisional data and 53,105 controls from ExAC (ALL) (OR 0.249, 95% CI 0.139-0.414, P = 2.86 × 10-11). PARP4 knockout did not affect the clonogenicity, proliferation rate, and migration of normal breast cells, but appeared to decrease the proliferation rate and clonogenicity of breast cancer cells. CONCLUSIONS: Taken together, our results do not support that PARP4 functions as a cancer susceptibility gene. This study highlights the importance of performing functional analyses for candidate cancer predisposition genes.


Asunto(s)
Biomarcadores de Tumor , Neoplasias de la Mama/genética , Predisposición Genética a la Enfermedad , Proteínas Nucleares/genética , Adulto , Anciano , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/mortalidad , Estudios de Casos y Controles , Línea Celular Tumoral , Proliferación Celular , Biología Computacional , Femenino , Técnicas de Silenciamiento del Gen , Estudios de Asociación Genética/métodos , Pruebas Genéticas , Heterocigoto , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Células Madre Neoplásicas/metabolismo , Medición de Riesgo , Factores de Riesgo , Singapur , Ensayo de Tumor de Célula Madre , Adulto Joven
12.
Sci Signal ; 11(554)2018 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-30377225

RESUMEN

RAS-RAF-MEK-ERK signaling has a well-defined role in cancer biology. Although aberrant pathway activation occurs mostly upstream of the kinase MEK, mutations in MEK are prevalent in some cancer subsets. Here, we found that cancer-related, activating mutations in MEK can be classified into two groups: those that relieve inhibitory interactions with the helix A region and those that are in-frame deletions of the ß3-αC loop, which enhance MEK1 homodimerization. The former, helix A-associated mutants, are inhibited by traditional MEK inhibitors. However, we found that the increased homodimerization associated with the loop-deletion mutants promoted intradimer cross-phosphorylation of the activation loop and conferred differential resistance to MEK inhibitors both in vitro and in vivo. MEK1 dimerization was required both for its activation by the kinase RAF and for its catalytic activity toward the kinase ERK. Our findings not only identify a previously unknown group of MEK mutants and provide insight into some key steps in RAF-MEK-ERK activation but also have implications for the design of therapies targeting RAS-ERK signaling in cancers.


Asunto(s)
Carcinogénesis , MAP Quinasa Quinasa 1/genética , Sistema de Señalización de MAP Quinasas , Neoplasias/genética , Animales , Transformación Celular Neoplásica , Fibroblastos/metabolismo , Células HEK293 , Humanos , MAP Quinasa Quinasa 1/antagonistas & inhibidores , Ratones , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Multimerización de Proteína , Eliminación de Secuencia , Transducción de Señal
13.
J Biol Chem ; 293(37): 14276-14284, 2018 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-30030377

RESUMEN

The dimerization-driven paradoxical activation of RAF proto-oncogene Ser/Thr kinase (RAF) is the predominant cause of drug resistance and toxicity in cancer therapies with RAF inhibitors. The scaffold protein 14-3-3, which binds to the RAF C terminus, is essential for RAF activation under physiological conditions, but the molecular basis is unclear. Here we investigated whether and how 14-3-3 regulates the dimerization-driven transactivation of the RAF isoform CRAF by RAF inhibitors and affects drug resistance and toxicity by virtue of the dominant role of CRAF in these processes. We demonstrated that 14-3-3 enhances the dimerization-driven transactivation of CRAF by stabilizing CRAF dimers. Further, we identified AMP-activated protein kinase (AMPK) and CRAF itself as two putative kinases that redundantly phosphorylate CRAF's C terminus and thereby control its association with 14-3-3. Next, we determined whether the combinatory inhibition of AMPK and CRAF could overcome the paradoxical effect of RAF inhibitors. We found that the AMPK inhibitor (AMPKi) not only blocked the RAF inhibitor-driven paradoxical activation of ERK signaling and cellular overgrowth in Ras-mutated cancer cells by blocking phosphorylation of Ser-621 in CRAF but also reduced the formation of drug-resistant clones of BRAFV600E-mutated cancer cells. Last, we investigated whether 14-3-3 binding to the C terminus of CRAF is required for CRAF catalytic activity and observed that it was dispensable in vivo Altogether, our study unravels the molecular mechanism by which 14-3-3 regulates dimerization-driven RAF activation and identified AMPKi as a potential agent to counteract drug resistance and adverse effects of RAF inhibitors in cancer therapies.


Asunto(s)
Adenilato Quinasa/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-raf/antagonistas & inhibidores , Serina/metabolismo , Proteínas 14-3-3/metabolismo , Línea Celular Tumoral , Dimerización , Células HEK293 , Humanos , Fosforilación , Unión Proteica , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-raf/química , Proteínas Proto-Oncogénicas c-raf/metabolismo , Serina/química , Transducción de Señal
14.
Oncogene ; 37(43): 5719-5734, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29930381

RESUMEN

Although extensively studied for three decades, the molecular mechanisms that regulate the RAF/MEK/ERK kinase cascade remain ambiguous. Recent studies identified the dimerization of RAF as a key event in the activation of this cascade. Here, we show that in-frame deletions in the ß3-αC loop activate ARAF as well as BRAF and other oncogenic kinases by enforcing homodimerization. By characterizing these RAF mutants, we find that ARAF has less allosteric and catalytic activity than the other two RAF isoforms, which arises from its non-canonical APE motif. Further, these RAF mutants exhibit a strong oncogenic potential, and a differential inhibitor resistance that correlates with their dimer affinity. Using these unique mutants, we demonstrate that active RAFs, including the BRAF(V600E) mutant, phosphorylate MEK in a dimer-dependent manner. This study characterizes a special category of oncogenic kinase mutations, and elucidates the molecular basis that underlies the differential ability of RAF isoforms to stimulate MEK-ERK pathway. Further, this study reveals a unique catalytic feature of RAF family kinases that can be exploited to control their activities for cancer therapies.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Mutación , Neoplasias , Multimerización de Proteína , Quinasas raf/metabolismo , Animales , Catálisis , Línea Celular Tumoral , Ratones , Ratones Noqueados , Neoplasias/enzimología , Neoplasias/genética , Neoplasias/patología , Quinasas raf/genética
15.
Virol J ; 13: 73, 2016 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-27129448

RESUMEN

BACKGROUND: Tiger frog virus (TFV), dsDNA virus of the genus Ranavirus and family Iridoviridae, causes a high mortality of tiger frog tadpoles cultured in Southern China. MicroRNAs (miRNAs) have been identified in many viruses especially DNA viruses such as Singapore Grouper Iridoviruses (SGIV). MicroRNAs play important roles in regulating gene expression for virus subsistence in host. Considering that TFV infects cells of different species under laboratory conditions, we aim to identify the specific and essential miRNAs expressed in ZF4 and HepG2 cells. METHODS: We identified and predicted novel viral miRNAs in TFV-infected ZF4 and HepG2 cells by deep sequencing and software prediction. Then, we verified and described the expression patterns of TFV-encoded miRNAs by using qRT-PCR and Northern blot. RESULTS: Deep sequencing predicted 24 novel TFV-encoded miRNAs, and qRT-PCR verified 19 and 23 miRNAs in TFV-infected ZF4 (Group Z) and HepG2 (Group H) cells, respectively. Northern blot was performed to validate eight and five TFV-encoded miRNAs in Groups H and Z, respectively. We compared the expression of TFV-encoded miRNAs from two groups and defined TFV-miR-11 as the essential viral miRNA and TFV-miR-13 and TFV-miR-14 as the specific miRNAs that contribute to HepG2 cell infection. CONCLUSIONS: We identified novel viral miRNAs and compared their expression in two host cells. The results of this study provide novel insights into the role of viral miRNAs in cross-species infection in vitro.


Asunto(s)
MicroARNs/análisis , ARN Viral/análisis , Ranavirus/crecimiento & desarrollo , Ranavirus/genética , Línea Celular , Biología Computacional , Perfilación de la Expresión Génica , Células Hep G2 , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , MicroARNs/genética , ARN Viral/genética , Análisis de Secuencia de ADN
16.
Fish Shellfish Immunol ; 45(2): 757-70, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26052019

RESUMEN

Cyprinid herpesvirus 3 (CyHV3) is a large double-stranded DNA virus of Alloherpesviridae family in the order Herpesvirales. It causes significant morbidity and mortality in common carp and its ornamental koi variety, and threatens the aquaculture industries worldwide. Mimicry of cytokines and cytokine receptors is a particular strategy for large DNA viruses in modulating the host immune response. Here, we report the identification and characterization of two novel viral homologues of tumor necrosis factor receptor (TNFR) encoded by CyHV3-ORF4 and -ORF12, respectively. CyHV3-ORF4 was identified as a homologue of HVEM and CyHV3-ORF12 as a homologue of TNFRSF1. Overexpression of ORF4 and ORF12 in zebrafish embryos results in embryonic lethality, morphological defects and increased apoptosis. Although we failed to identify any interaction between the two vTNFRs and their potential ligands in zebrafish TNF superfamily by yeast two-hybrid system, the expression of some genes in TNF superfamily or TNFR superfamily were mis-regulated in ORF4 or ORF12-overexpressing embryos, especially the death receptor zHDR and its cognate ligand DL1b. Further studies showed that the apoptosis induced by the both CyHV3 vTNFRs is mainly activated through the intrinsic apoptotic pathway and requires the crosstalk between the intrinsic and extrinsic apoptotic pathway. Additionally, using RT-qPCR and Western blot assays, the expression patterns of the both vTNFRs were also analyzed during CyHV3 productive infection. Collectively, this is the first functional study of two unique vTNFRs encoded by a herpesvirus infecting non-mammalian vertebrates, which may provide novel insights into viral immune regulation mechanism and the pathogenesis of CyHV3 infection.


Asunto(s)
Enfermedades de los Peces/genética , Infecciones por Herpesviridae/veterinaria , Herpesviridae/fisiología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Proteínas Virales/genética , Pez Cebra , Secuencia de Aminoácidos , Animales , Carpas , Línea Celular , Femenino , Enfermedades de los Peces/metabolismo , Enfermedades de los Peces/virología , Regulación de la Expresión Génica , Herpesviridae/genética , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/metabolismo , Infecciones por Herpesviridae/virología , Masculino , Sistemas de Lectura Abierta , Miembro 14 de Receptores del Factor de Necrosis Tumoral/química , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/química , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Alineación de Secuencia/veterinaria , Proteínas Virales/química , Proteínas Virales/metabolismo
17.
J Virol ; 89(1): 763-75, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25355883

RESUMEN

UNLABELLED: Infectious spleen and kidney necrosis virus (ISKNV) is the type species of the Megalocytivirus genus, Iridoviridae family, causing a severe systemic disease with high mortality in mandarin fish (Siniperca chuatsi) in China and Southeast Asia. At present, the pathogenesis of ISKNV infection is still not fully understood. Based on a genome-wide bioinformatics analysis of ISKNV-encoded proteins, we found that ISKNV open reading frame 119L (ORF119L) is predicted to encode a three-ankyrin-repeat (3ANK)-domain-containing protein, which shows high similarity to the dominant negative form of integrin-linked kinase (ILK); i.e., viral ORF119L lacks the ILK kinase domain. Thus, we speculated that viral ORF119L might affect the host ILK complex. Here, we demonstrated that viral ORF119L directly interacts with particularly interesting Cys-His-rich protein (PINCH) and affects the host ILK-PINCH interaction in vitro in fathead minnow (FHM) cells. In vivo ORF119L overexpression in zebrafish (Danio rerio) embryos resulted in myocardial dysfunctions with disintegration of the sarcomeric Z disk. Importantly, ORF119L overexpression in zebrafish highly resembles the phenotype of endogenous ILK inhibition, either by overexpressing a dominant negative form of ILK or by injecting an ILK antisense morpholino oligonucleotide. Intriguingly, ISKNV-infected mandarin fish develop disorganized sarcomeric Z disks in cardiomyocytes. Furthermore, phosphorylation of AKT, a downstream effector of ILK, was remarkably decreased in ORF119L-overexpressing zebrafish embryos. With these results, we show that ISKNV ORF119L acts as a domain-negative inhibitor of the host ILK, providing a novel mechanism for the megalocytivirus pathogenesis. IMPORTANCE: Our work is the first to show the role of a dominant negative inhibitor of the host ILK from ISKNV (an iridovirus). Mechanistically, the viral ORF119L directly binds to the host PINCH, attenuates the host PINCH-ILK interaction, and thus impairs ILK signaling. Intriguingly, ORF119L-overexpressing zebrafish embryos and ISKNV-infected mandarin fish develop similar disordered sarcomeric Z disks in cardiomyocytes. These findings provide a novel mechanism for megalocytivirus pathogenesis.


Asunto(s)
Interacciones Huésped-Patógeno , Iridoviridae/fisiología , Proteínas Musculares/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Virales/metabolismo , Proteínas de Pez Cebra/antagonistas & inhibidores , Proteínas de Pez Cebra/metabolismo , Animales , Línea Celular , Cyprinidae , Infecciones por Virus ADN/patología , Infecciones por Virus ADN/virología , Modelos Animales de Enfermedad , Miocardio/patología
18.
PLoS One ; 7(5): e37001, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22615868

RESUMEN

Infectious spleen and kidney necrosis virus (ISKNV) is the type species of the Megalocytivirus genus of the Iridoviridae family. It causes a serious and potentially pandemic disease in wild and cultured fishes. ISKNV infection induces evident apoptosis in mandarin fish (Siniperca chuatsi) and zebrafish (Danio renio). However, the mechanism is still unknown. After a genome-wide bioinformatics analysis of ISKNV-encoded proteins, the ISKNV open reading frame 111L (ORF111L) shows a high similarity to the tumour necrosis factor receptor-associated factor (TRAF) encoded by fish, mice and mammals, which is essential for apoptotic signal transduction. Moreover, ORF111L was verified to directly interact with the zebrafish TNF receptor type 1 associated death domain protein (TRADD). A recombinant plasmid containing the DNA sequence of ORF111L was constructed and microinjected into zebrafish embryos at the 1-2 cell stage to investigate its biological function in vivo. ORF111L overexpression in the embryos resulted in increased apoptosis. ORF111L-induced apoptosis was clearly associated with significant caspase 8 upregulation and activation. The knockdown of zebrafish caspase 8 expression effectively blocked the apoptosis induced by ORF111L overexpression. Significantly, ORF111L overexpression resulted in much stronger effect on caspase 8 and caspase 3 upregulation compared to zebrafish TRAF2. This is the first report of a viral protein similar to TRAF that interacts with TRADD and induces caspase 8-mediated apoptosis, which may provide novel insights into the pathogenesis of ISKNV infection.


Asunto(s)
Apoptosis/fisiología , Caspasa 8/metabolismo , Iridoviridae/metabolismo , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/genética , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo , Animales , Caspasa 3/genética , Caspasa 3/metabolismo , Caspasa 8/genética , Biología Computacional/métodos , ADN Viral/genética , Enfermedades de los Peces/genética , Enfermedades de los Peces/metabolismo , Enfermedades de los Peces/virología , Iridoviridae/genética , Sistemas de Lectura Abierta , Proteína de Dominio de Muerte Asociada a Receptor de TNF/genética , Proteína de Dominio de Muerte Asociada a Receptor de TNF/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Pez Cebra
19.
J Clin Lab Anal ; 21(6): 398-405, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18022924

RESUMEN

Pleural effusions (PE) are the most common complications that may be produced by a wide variety of diseases. A large number of studies exploring the role of carcinoembryonic antigen (CEA) and cytokeratin fragment 19 (CYFRA 21-1) marker in differential diagnosis of PE have been published, employing differing methodologies with sometimes conflicting results. A comprehensive systematic review would be useful to synthesize the currently available bulk of information. The objective of this work was to assess and compare the overall value of pleural fluid CEA and CYFRA 21-1 in differential diagnosis of PEs with a meta-analysis. All the English and Chinese published studies for differential diagnosis of PEs by pleural fluid CEA and CYFRA 21-1 were collected. Methodological quality of the included studies was evaluated. Pooled sensitivity and specificity were calculated, the threshold effect and the possible sources of heterogeneity were also analyzed. Summary receiver operating characteristic (SROC) curve analysis was used to compare the differential diagnostic ability of pleural fluid CEA and CYFRA 21-1. A total of 19 studies were included in the meta-analysis, with a total of 3,228 subjects. Pooled sensitivity and specificity of CEA and CYFRA 21-1 were 45.9% (43.2-48.5%) and 97.0% (96.0-97.8%), and 47.3% (44.0-50.6%) and 91.8% (89.5-93.7%), respectively. Both CEA and CYFRA 21-1 have a threshold effect, the main source of heterogeneity was from variable assay methods. The areas under the SROC curve (AUCs) of CEA and CYFRA 21-1 were 0.7691 and 0.8213, respectively. There was no statistical significance between the AUC of CEA and CYFRA 21-1 (P>0.05). Both CEA and CYFRA 21-1 have good performance in the differential diagnosis of PE, when compared with CEA, CYFRA 21-1 has no advantage.


Asunto(s)
Antígenos de Neoplasias/análisis , Antígeno Carcinoembrionario/análisis , Queratinas/análisis , Derrame Pleural Maligno/inmunología , Humanos , Queratina-19 , Derrame Pleural Maligno/diagnóstico , Sensibilidad y Especificidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...