Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
FASEB J ; 36(6): e22341, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35579877

RESUMEN

Multiple myeloma (MM), the terminally differentiated B cells malignancy, is widely considered to be incurable since many patients have either developed drug resistance or experienced an eventual relapse. To develop precise and efficient therapeutic strategies, we must understand the pathogenesis of MM. Thus, unveiling the driver events of MM and its further clonal evolution will help us understand this complicated disease. Chromosome 1 instabilities are the most common genomic alterations that participate in MM pathogenesis, and these aberrations of chromosome 1 mainly include copy number variations and structural changes. The chromosome 1q gains/amplifications and 1p deletions are the most frequent structural changes of chromosomes in MM. In this review, we intend to focus on the genes that are affected by chromosome 1 instability: some tumor suppressors were lost or down regulated in 1p deletions, and others that contributed to tumorigenesis were upregulated in 1q gains/amplifications. We have summarized their biological function as well as their roles in the MM pathogenesis, hoping to uncover potential novel therapeutical targets and promote the development of future therapeutic approaches.


Asunto(s)
Mieloma Múltiple , Inestabilidad Cromosómica , Aberraciones Cromosómicas , Cromosomas Humanos Par 1/genética , Variaciones en el Número de Copia de ADN , Expresión Génica , Humanos , Mieloma Múltiple/genética , Mieloma Múltiple/terapia
2.
Parasite Immunol ; 43(8): e12842, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33959966

RESUMEN

AIMS: Schistosomiasis is a parasitic disease with a chronic debilitating character caused by parasitic flatworms of the genus Schistosoma. The main disease-causing species of Schistosoma in China is S. japonicum. M fortis has been proved to be a nonpermissive host of S. japonicum. Mf-HSP90α (Microtus fortis heat shock protein 90alpha), the homologue of HSP90α, display anti-schistosome effect in vitro and in vivo. In the current study, in order to investigate the mechanism of anti-schistosome effect of Mf-HSP90α, we conducted RNA-Seq to obtain the transcriptome profile of M. fortis liver infected with S. japonicum at different time points. METHODS AND RESULTS: By mapping the differential expressed genes (DEGs) to Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG), we found that the JAK2/STAT1 pathway was highly enriched with an elevated level of IL-10 and HSP90α. We then checked the IL-10-JAK2/STAT1-HSP90α pathway, and found that this pathway was activated in the infected mice with S. japonicum. The expression of the molecules in this pathway was elevated on the 10th day after infection and gradually decreased on the 20th day. CONCLUSIONS: The IL-10-JAK2/STAT1-HSP90α axis was associated with the anti-schistosome effect of Mf-HSP90α, and targeting IL-10-JAK2/STAT1-HSP90α axis might be a novel therapeutic strategy for developing resistance to S. japonicum infection.


Asunto(s)
Schistosoma japonicum , Esquistosomiasis Japónica , Esquistosomiasis , Animales , Arvicolinae , Hígado , Ratones , Transcriptoma
3.
Biotechnol Lett ; 43(3): 537-546, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33386501

RESUMEN

OBJECTIVE: Two-dimensional electrophoresis (2-DE) and MALDI-TOF/TOF mass spectrometry were performed to compare the proteomic alterations of lycorine-treated and control cells to further investigate the anti-multiple myeloma (MM) mechanisms of lycorine. RESULTS: Mass spectrometry results showed that after lycorine treatment of MM cells, 42% of the differentially expressed proteins had subcellular localization, mainly, on mitochondria. Voltage-dependent anion-selective channel protein 2 (VDAC2), the most abundant protein in the outer mitochondrial membrane, was up-regulated after treatment with lycorine and was subsequently verified by western blot analysis. Further studies on mitochondria found that lycorine was able to increase abnormal mitochondria and increase mitochondrial membrane potential. CONCLUSIONS: Lycorine can achieve the effect of resisting multiple myeloma by acting on VDAC2 and causing mitochondrial abnormalities.


Asunto(s)
Alcaloides de Amaryllidaceae/farmacología , Mieloma Múltiple/metabolismo , Fenantridinas/farmacología , Proteoma/efectos de los fármacos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Canal Aniónico 2 Dependiente del Voltaje/metabolismo , Antineoplásicos/farmacología , Electroforesis en Gel Bidimensional , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Proteoma/análisis
4.
Br J Haematol ; 191(1): 62-76, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32314355

RESUMEN

Multiple myeloma stem-like cells (MMSCs) are responsible for initiation and relapse, though novel treatment paradigms that effectively eradicate MMSCs are yet to be developed. Selective inhibition of the cell cycle regulatory kinase Wee1 by MK1775 is being explored as a potential anti-cancer therapeutic. We report that higher expression of Wee1 is correlated with poor survival in multiple myeloma (MM). The MM models and patient-derived CD138+ plasma cells are particularly sensitive to the growth-inhibitory effects of the Wee1 inhibitor MK1775. MK1775 induces Mus81-Eme1 endonuclease-mediated DNA damage in S-phase cell cycle that results in a blockade of replication and then apoptosis. Furthermore, MK1775 strongly suppresses the features of stemness in vitro, in vivo and in primary CD138+ cells by decreasing ALDH1+ cell fraction and the expression of ALDH1. In addition, co-treatment of MK1775 with bortezomib is synergistic in vitro and in vivo. Bortezomib, although it enhances ALDH1+ cells, when combined with MK1775 abrogates this stimulatory effect on stemness. Considering MM as an invariably incurable malignancy due to the presence of heterogenic myeloma stem-like cells, our study presents inhibition of Wee1 as a promising targeted therapy for MM and provides a compelling rationale to further investigate the activity of MK1775 against myeloma in clinical settings.


Asunto(s)
Bortezomib/farmacología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Mieloma Múltiple , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirazoles/farmacología , Pirimidinonas/farmacología , Animales , Bortezomib/agonistas , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Sinergismo Farmacológico , Humanos , Ratones , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/enzimología , Proteínas de Neoplasias/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Pirazoles/agonistas , Pirimidinonas/agonistas , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Artículo en Zh | MEDLINE | ID: mdl-30146861

RESUMEN

Objective: To investigate the resistance of E77.43 gene of Microtus fortis(MfE77.43) to Schistosoma japonicum infection. Methods: MfE77.43 was constructed into the recombinant Adeno-associated virus AAV2. The AAV2-MfE77.43 was transfected into HEK293 cells by the calcium phosphate DNA coprecipitation method. The recombinant rAAV2-MfE77.43 was purified and total RNA was extracted from the transfected cells. The expression of E77.43 was examined by RT-PCR and the purity of rAAV2-MfE77.43 was analyzed by SDS-PAGE. Eighteen KM mice were divided into three groups (n=6 in each group). Mice in the experiment group were intramuscularly injected on days 0, 3 and 7 with 400 µl recombinant AAV2-MfE77.43 virus which was 5-fold diluted in normal saline. Mice in negative control and blank control groups received same volume of pAAV or normal saline. Venous blood was collected through the tail before each injection, and E77.43 expression in plasma was detected by dot-ELISA method. After the last injection, each mouse was infected with 40 S. japonicum cercariae and sacrificed on day 41 after infection. Adult worms and liver eggs per gram(LEPG) were counted. Worm and egg reduction rate was calculated respectively. Egg granulomas were observed by HE staining. Results: RT-PCR resulted in a 330 bp specific band. SDS-PAGE of virus shell protein revealed three protein bands with M(r) of 87 000, 72 000 and 62 000, respectively. Dot-ELISA showed that E77.43 protein began to be expressed on day 3 after rAAV2-MfE77.43 injection, remaining stable till day 41. The adult worm number and LEPG were 20.16±3.93 and 19 800±2 715, respectively, with a worm and egg reduction rate of 27.3% and 26.2% in the experiment group. While the worm number and LEPG in the negative control group were 29.16±2.44 and 28 000±2 192(P<0.01), respectively. HE staining and observation revealed fewer eosinophils and inflammatory cells around the liver eggs in the therapy group. Conclusion: The E77.43 gene shows protective effects against S. japonicum infection, indicating that E77.43 may participate in the natural resistance of Microtus fortis to S. japonicum infection.


Asunto(s)
Schistosoma japonicum , Esquistosomiasis Japónica , Animales , Arvicolinae , Cercarias , Granuloma , Células HEK293 , Humanos , Hígado , Ratones , Ratones Endogámicos BALB C
6.
Neoplasia ; 50: 100974, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38364355

RESUMEN

Multiple myeloma is a hematological malignancy that has evolved from antibody-secreting B lymphocytes. Like other types of cancers, myeloma cells have acquired functional capabilities which are referred to as "Hallmarks of Cancer", and one of their most important features is the metabolic disorders. Due to the high secretory load of the MM cells, the first-line medicine proteasome inhibitors have found their pronounced effects in MM cells for blocking the degradation of misfolded proteins, leading to their accumulation in the ER and overwhelming ER stress. Moreover, proteasome inhibitors have been reported to be effective in myeloma by targeting glucose, lipid, amino acid metabolism of MM cells. In this review, we have described the abnormal metabolism of the three major nutrients, such as glucose, lipid and amino acids, which participate in the cellular functions. We have described their roles in myeloma progression, how they could be exploited for therapeutic purposes, and current therapeutic strategies targeting these metabolites, hoping to uncover potential novel therapeutic targets and promote the development of future therapeutic approaches.


Asunto(s)
Mieloma Múltiple , Inhibidores de Proteasoma , Humanos , Inhibidores de Proteasoma/farmacología , Inhibidores de Proteasoma/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/metabolismo , Glucosa , Lípidos/uso terapéutico , Complejo de la Endopetidasa Proteasomal/metabolismo
7.
Cancer Lett ; 554: 216019, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36442773

RESUMEN

Multiple myeloma (MM) is an incurable condition and the second most common hematological malignancy. Over the past few years, there has been progress in the treatment of MM, but most patients still relapse. Multiple myeloma stem-like cells (MMSCs) are believed to be the main reason for drug resistance and eventual relapse. Currently, there are not enough therapeutic agents that have been identified for eradication of MMSCs, and thus, identification of the same may alleviate the issue of relapse in patients. In the present study, we showed that luteolin (LUT), a natural compound obtained from different plants, such as vegetables, medicinal herbs, and fruits, effectively inhibits the proliferation of MM cells and overcomes bortezomib (BTZ) resistance in them in vitro and in vivo, mainly by decreasing the proportion of ALDH1+ cells. Furthermore, RNA sequencing after LUT treatment of MM cell lines and an MM xenograft mouse model revealed that the effects of the compound are mediated through inhibition of transforming growth factor-ß signaling. Similarly, we found that LUT also significantly reduced the proportion of ALDH1+ cells in primary CD138+ plasma cells. In addition, LUT could overcome the BTZ treatment-induced increase in the proportion of ALDH1+ cells, and the combination of LUT and BTZ had a synergistic effect against myeloma cells. Collectively, our findings suggested that LUT is a promising agent that manifests MMSCs to overcome BTZ resistance, alone or in combination with BTZ, and thus, is a potential therapeutic drug for the treatment of MM.


Asunto(s)
Antineoplásicos , Mieloma Múltiple , Humanos , Animales , Ratones , Bortezomib/farmacología , Mieloma Múltiple/patología , Luteolina/farmacología , Resistencia a Antineoplásicos , Apoptosis , Línea Celular Tumoral , Recurrencia Local de Neoplasia/tratamiento farmacológico , Transducción de Señal , Factor de Crecimiento Transformador beta/farmacología , Antineoplásicos/farmacología
8.
Cancer Lett ; 537: 215647, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35306105

RESUMEN

Multiple myeloma is a plasma cell malignancy, accounting for approximately 1% of new cancer cases. It is the second most common hematological malignancy. Novel clinical agents such as the proteasome inhibitor-bortezomib, have shown improved survival rates in recent decades. However, multiple myeloma remains incurable, as most patients eventually relapse and become refractory to current treatments. Therefore, there is an urgent need for developing new regimens to overcome the bortezomib resistance. Here, we screened a library of 2370 bioactives and found that polyphyllin VII selectively suppressed multiple myeloma cell growth in vitro and in vivo. We identified moesin, one of the critical regulators of the Wnt/ß-catenin pathway, as a target of polyphyllin VII by drug affinity responsive target stability assay and cellular thermal shift assay. Polyphyllin VII binds to moesin and induces its degradation via the ubiquitin-proteasome pathway, thereby impairing the Wnt/ß-catenin pathway activity and leading to a reduction in the side population cells to overcome bortezomib resistance. Our study identified polyphyllin VII as a promising compound and moesin as a potential diagnostic and therapeutic target for treating multiple myeloma.


Asunto(s)
Antineoplásicos , Mieloma Múltiple , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Bortezomib/farmacología , Bortezomib/uso terapéutico , Línea Celular Tumoral , Resistencia a Antineoplásicos , Humanos , Proteínas de Microfilamentos , Mieloma Múltiple/patología , Recurrencia Local de Neoplasia/tratamiento farmacológico , Inhibidores de Proteasoma/farmacología , Inhibidores de Proteasoma/uso terapéutico , Saponinas , beta Catenina/metabolismo
9.
Tumour Biol ; 32(6): 1133-8, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21811875

RESUMEN

To investigate the effects of methylation of the p73 gene on the pathogenesis of non-Hodgkin lymphoma (NHL), the methylation status of the p73 gene promoter and the expression of p73 mRNA were examined in NHLs by methylation-specific polymerase chain reaction (MSP) and reverse transcription-polymerase chain reaction, respectively; p73 protein was detected by Western blotting analysis. Furthermore, the expression of p73 mRNA in NHL cells treated with 5-Aza-2'-deoxycytidine was analyzed. MSP results revealed that the promoter of p73 was methylated in 87.5% of NHLs but was not methylated in reactive hyperplasia lymph node samples. The expression of p73 mRNA was not detected in 83.33% of NHLs but was detected in all of the reactive hyperplasia lymph node samples. The p73 protein was not detected in 91.67% of NHLs but was detected in all of the reactive hyperplasia lymph node samples. The expression of p73 mRNA was detected in NHL cells treated with 5-Aza-2'-deoxycytidine. The inactivation of p73, predominantly by methylation, may be involved in the pathogenesis of NHLs.


Asunto(s)
Metilación de ADN , Proteínas de Unión al ADN/genética , Linfoma no Hodgkin/genética , Proteínas Nucleares/genética , Proteínas Supresoras de Tumor/genética , Adulto , Antimetabolitos Antineoplásicos/farmacología , Azacitidina/análogos & derivados , Azacitidina/farmacología , Western Blotting , Proteínas de Unión al ADN/metabolismo , Decitabina , Relación Dosis-Respuesta a Droga , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Linfoma no Hodgkin/metabolismo , Linfoma no Hodgkin/patología , Masculino , Persona de Mediana Edad , Proteínas Nucleares/metabolismo , Reacción en Cadena de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Proteína Tumoral p73 , Proteínas Supresoras de Tumor/metabolismo , Adulto Joven
10.
Cancer Lett ; 520: 307-320, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34390764

RESUMEN

Multiple myeloma (MM) is incurable and the second most common hematologic malignancy in plasma cells. Multiple myeloma stem cell-like cells (MMSCs), a rare population of MM cells, are believed to be the major cause of drug resistance and high recurrence rates in patients with MM. Therefore, developing novel strategies to eradicate MMSCs may favor myeloma treatment. In this study, based on the drug repositioning strategy, we found that albendazole (ABZ), a broad-spectrum antiparasitic drug, selectively suppresses the proliferation of multiple myeloma cells in vitro and in vivo and decreases number of aldehyde dehydrogenase (ALDH)-positive MMSCs in MM. Furthermore, RNA-seq of MM cells after ABZ treatment revealed that inhibition of the nuclear factor kappa-B (NF-κB) pathway is a key mediator of ABZ against MM. Moreover, we demonstrated that ABZ can resensitize cells resistant to bortezomib and overcome MMSCs-induced bortezomib resistance by decreasing ALDH1+ MMSCs numbers. Our findings provide preclinical evidence for utilizing the previously known pharmacologically active drug albendazole for the treatment of multiple myeloma.


Asunto(s)
Albendazol/farmacología , Bortezomib/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Mieloma Múltiple/tratamiento farmacológico , Familia de Aldehído Deshidrogenasa 1/genética , Protocolos de Quimioterapia Combinada Antineoplásica , Apoptosis/efectos de los fármacos , Bortezomib/efectos adversos , Línea Celular Tumoral , Humanos , Mieloma Múltiple/genética , FN-kappa B/genética , Células Madre Neoplásicas/efectos de los fármacos , RNA-Seq , Transducción de Señal/efectos de los fármacos
11.
Biomed Res Int ; 2020: 6960497, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32090108

RESUMEN

OBJECTIVE: This study aimed to evaluate the potential effect of hyperhomocysteinemia on multiple organ failure (MOF) in patients with acute pancreatitis (AP). METHOD: In this cohort study, a total of 1880 AP patients were enrolled and divided into the hyperhomocysteinemia group (study group) and the control group based on serum homocysteine (HCY) levels. Clinical data including demographics, clinical outcomes, and characteristics were collected for analysis. Risk factors of MOF in AP patients were determined by univariate and multivariate logistic regression analyses. RESULTS: The hyperhomocysteinemia group showed higher multiple organ failure rates (31.83% vs 20.77%, P < 0.001), compared with the control group. A positive correlation between homocysteine level and APACHE II score was obtained by Pearson correlation analysis (r = 0.420, P < 0.001), compared with the control group. A positive correlation between homocysteine level and APACHE II score was obtained by Pearson correlation analysis (P < 0.001), compared with the control group. A positive correlation between homocysteine level and APACHE II score was obtained by Pearson correlation analysis (. CONCLUSION: A high serum homocysteine level may be an independent risk factor of multiple organ failure in patients with acute pancreatitis.


Asunto(s)
Hiperhomocisteinemia/complicaciones , Insuficiencia Multiorgánica/complicaciones , Pancreatitis/complicaciones , Proteína C-Reactiva/metabolismo , Estudios de Casos y Controles , Femenino , Homocisteína/sangre , Humanos , Hiperhomocisteinemia/sangre , Recuento de Leucocitos , Modelos Logísticos , Masculino , Persona de Mediana Edad , Insuficiencia Multiorgánica/sangre , Análisis Multivariante , Pancreatitis/sangre , Curva ROC , Índice de Severidad de la Enfermedad , Resultado del Tratamiento
12.
Oncogene ; 39(16): 3354-3366, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32103167

RESUMEN

Multiple myeloma (MM) is a clinically and biologically heterogenous event that accounts for approximately 10% of all hematological malignancies. Chromosome 1 open reading frame 35 (C1orf35) is a gene cloned and identified in our laboratory from a MM cell line (GenBank: AY137773), but little is known about its function. In the current study, we have confirmed that C1orf35 is a candidate oncogene, and it can promote cell cycle progression from G1 to S. Later, we found that C1orf35 is able to affect the cell proliferation by modulating the expression of c-MYC (v-myc myelocytomatosis viral oncogene homolog), and the oncogenic property of C1orf35 can be rescued by c-MYC inhibition. Herein, we found positive association between C1orf35 and c-MYC in MM patients and in MM cell lines. The correlation analysis of the genes coamplified in MM patients from GEO datasets showed a correlation between C1orf35 and c-MYC, and the expression data of different stages of plasma cell neoplasm acquired from GEO datasets showed that the expression of C1orf35 increase with the progression of the disease. This indicates that C1orf35 may play a role in the disease progression. Moreover, C1orf35 can modulate c-MYC expression and rescue c-MYC transcription inhibited by Act D. Finally, we have shown that C1orf35 activates c-MYC transcription by binding to the i-motif of Nuclease hypersensitivity element III1 (NHE III1) in the c-MYC promoter. Not only does our current study advance our knowledge of the pathogenesis and therapeutic landscape of MM, but also of other cancer types and diseases that are initiated with deregulated c-MYC transcription.


Asunto(s)
Carcinogénesis/genética , Mieloma Múltiple/genética , Proteínas de Neoplasias/genética , Proteínas Proto-Oncogénicas c-myc/genética , Adulto , Anciano , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Mieloma Múltiple/patología , Células 3T3 NIH , Regiones Promotoras Genéticas/genética , Unión Proteica/genética , Transcripción Genética/genética , Activación Transcripcional/genética
13.
Cell Signal ; 60: 136-145, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31034872

RESUMEN

Multiple myeloma (MM) is hematological malignancy characterized by clonal proliferation of malignant plasma cells in the bone marrow environment. Previously, we identified DAZAP2 as a candidate cancer suppressor gene, the downregulation of which is regulated by its own promoter methylation status. In the current study, we analyzed the DAZAP2 promoter in MM cell lines KM3, MM.1S, OPM-2, and ARH77 by bisulfite genomic sequencing assay. We identified the binding site for transcription factor cyclic adenosine monophosphate response element binding (CREB) in the DAZAP2 promoter CpG2, and we found that hypermethylation of the CREB binding motif in the DAZAP2 promoter is responsible for the reduced DAZAP2 expression in MM cells. Later we checked the p38/MAPK signaling cascade, which is reported to regulate expression and function of CREB. Our results showed that the p38/MAPK signaling pathway drives the expression of DAZAP2 by phosphorylation of CREB, and hypermethylation of CREB binding motif in DAZAP2 promoter can inhibit binding of CREB to the latter, thus downregulating DAZAP2 expression. Moreover, treating the MM cells with 5-aza-2' deoxycytidine to demethylate DAZAP2 promoter restored the binding of CREB to its binding motif, and thus upregulated DAZAP2 expression. Our results not only identified DAZAP2 as a new downstream target of p38/MAPK/CREB signaling cascade, but we also clarified that the downregulation of DAZAP2 in MM cells is caused by hypermethylation of CREB binding motif in its own promoter region, which implies that demethylation of DAZAP2 promoter can be a novel therapeutic strategy for MM treatment.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Mieloma Múltiple/metabolismo , Proteínas de Unión al ARN/genética , Línea Celular Tumoral , Metilación de ADN , Humanos , Regiones Promotoras Genéticas
14.
BMC Cancer ; 6: 66, 2006 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-16539746

RESUMEN

BACKGROUND: HSVtk/ganciclovir (GCV) gene therapy has been extensively studied in tumors and relies largely on the gene expression of HSVtk. Most studies, however, have failed to demonstrate any significant benefit of a controlled gene expression strategy in cancer treatment. The Tet-On system is commonly used to regulate gene expression following Dox induction. We have evaluated the antitumor effect of HSVtk/ganciclovir gene therapy under Tet-On regulation by means of adeno-associated virus-2 (AAV-2)-mediated HSVtk gene transfer with direct intratumoral injections in mice bearing breast cancer tumors. METHODS: Recombinant adeno-associated virus-2 (rAAV) was constructed and transduced into MCF-7 cell line. GCV treatment to the rAAV infected MCF-7 cells was performed by MTT assay under the doxycycline (Dox) induction or without Dox induction at a vp (viral particle) number of > or =10(4)/cell. The virus was administered intratumorally to nude mice that had also received GCV intraperitoneally. The antitumor effects were evaluated by measuring tumor regression and histological analysis. RESULTS: We have demonstrated that GCV treatment to the infected MCF-7 cells under the Dox induction was of more inhibited effects than those without Dox induction at > or =10(4) vp/cell. In ex vivo experiments, tumor growth of BALB/C nude mice breast cancer was retarded after rAAV-2/HSVtk/Tet-On was injected into the tumors under the Dox induction. Infiltrating cells were also observed in tumors after Dox induction followed by GCV treatment and cells were profoundly damaged. The expression of HSVtk gene in MCF-7 cells and BALB/C nude mice tumors was up-regulated by Tet-On under Dox induction with reverse transcription-PCR (RT-PCR) analysis. CONCLUSION: The antitumor effect of rAAV-mediated HSVtk/GCV gene therapy under the Dox induction with direct intratumoral injections may be a useful treatment for breast cancer and other solid tumors.


Asunto(s)
Adenocarcinoma/terapia , Neoplasias de la Mama/terapia , Dependovirus/genética , Genes Transgénicos Suicidas , Terapia Genética , Vectores Genéticos/uso terapéutico , Simplexvirus/enzimología , Timidina Quinasa/genética , Adenocarcinoma/patología , Animales , Antivirales/uso terapéutico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Doxiciclina/farmacología , Inducción Enzimática , Femenino , Ganciclovir/uso terapéutico , Regulación Viral de la Expresión Génica/efectos de los fármacos , Genes Sintéticos , Vectores Genéticos/administración & dosificación , Humanos , Inyecciones Intralesiones , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Secuencias Reguladoras de Ácidos Nucleicos , Timidina Quinasa/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Artículo en Zh | MEDLINE | ID: mdl-16566206

RESUMEN

OBJECTIVE: To explore the killing effect of different fractional proteins from Microtus fortis (Mf) serum to S. japonicum juveniles, and to find possible association of the proteins with the natural resistance to schistosome infection. METHODS: The proteins in Mf serum were separated by means of ion-exchange column chromatography and molecular sieve column chromatography. After desalted by dialysis and lyophilized, the proteins were dissolved in DMEM medium which contained 300 U/ml penicillin and 300 microg/ml streptomycin, and the two-day old schistosomula were added in for in vitro cultivation (100 +/- 20/well). The killing activity of the fractional proteins to the juvenile worms was defined by mortality rate. RESULTS: 58 fractional proteins were separated from Mf serum, in which six proteins were confirmed to have a significant killing activity to schistosomula. The mortality of schistosomula all reached 37% and above, and the highest mortality (87.5%) was observed in the fraction 18.1, while the negative control was 25.09% (P < 0.01). CONCLUSION: Some fractional proteins in Microtus fortis serum show an effect in the natural resistance to schistosome infection.


Asunto(s)
Arvicolinae/inmunología , Proteínas Sanguíneas/farmacología , Schistosoma japonicum/crecimiento & desarrollo , Animales , Proteínas Sanguíneas/aislamiento & purificación , Medios de Cultivo , Larva/efectos de los fármacos , Larva/crecimiento & desarrollo , Ratones , Ratones Endogámicos , Conejos , Schistosoma japonicum/efectos de los fármacos
16.
Genomics Proteomics Bioinformatics ; 2(1): 47-54, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15629043

RESUMEN

In our previous studies, DAZAP2 gene expression was down-regulated in untreated patients of multiple myeloma (MM). For better studying the structure and function of DAZAP2, a full-length cDNA was isolated from mononuclear cells of a normal human bone marrow, sequenced and deposited to Genbank (AY430097). This sequence has an identical ORF (open reading frame) as the NM_014764 from human testis and the D31767 from human cell line KG-1. Phylogenetic analysis and structure prediction reveal that DAZAP2 homologues are highly conserved throughout evolution and share a polyproline region and several potential SH2/SH3 binding sites. DAZAP2 occurs as a single-copy gene with a four-exon organization. We further noticed that the functional DAZAP2 gene is located on Chromosome 12 and its pseudogene gene is on Chromosome 2 with electronic location of human chromosome in Genbank, though no genetic abnormalities of MM have been reported on Chromosome 12. The ORF of human DAZAP2 encodes a 17-kDa protein, which is highly similar to mouse Prtb. The DAZAP2 protein is mainly localized in cytoplasm with a discrete pattern of punctuated distribution. DAZAP2 may associate with carcinogenesis of MM and participate in yet-to-be identified signaling pathways to regulate proliferation and differentiation of plasma cells.


Asunto(s)
Cromosomas Humanos Par 12/genética , Regulación hacia Abajo , Mieloma Múltiple/genética , Filogenia , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Cromosomas Humanos Par 2/genética , Citoplasma/metabolismo , Cartilla de ADN , ADN Complementario/genética , Componentes del Gen , Humanos , Funciones de Verosimilitud , Modelos Genéticos , Datos de Secuencia Molecular , Mieloma Múltiple/metabolismo , Seudogenes/genética , Alineación de Secuencia , Análisis de Secuencia de ADN
17.
Artículo en Inglés | MEDLINE | ID: mdl-12058174

RESUMEN

Human stem cell factor(hSCF)is a pluripotent growth factor that regulates proliferation, differentiation and migration of certain mammalian stem cells, such as primordial germ cells etc. It is shown that hSCF and its receptor are commonly co-expressed in human breast cancer cells. Up to now, the definite regulatory mechanism of hSCF gene in breast cancer cells is unclear, except that its 5'flanking sequence contains essential elements for regulating transcription. To localize the regulatory elements responsible for the regulation of the hSCF gene, we performed transient transfection study in MCF cells, with a series of luciferase reporter gene constructs, containing different 5x end deletions of hSCF gene. This study indicates that the region of -1190 -853 significantly enhanced the luc gene expression, while the region of -339 -162 inhibited the expression. Eletrophoretic mobility shift assay confirmed that MCF nuclear extract proteins bound to both -1190 -853 and -339 -273 regions, forming specific DNA-protein complexes, indicating that there were nuclear protein binding sites in these regions. The results suggest that both -1190 -853 and -339 -273 DNA fragments of the hSCF 5'flanking sequence may be novel regulatory elements, and may play a role in the regulation of hSCF gene expression in MCF cells.

18.
Chin Med J (Engl) ; 117(3): 434-9, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15043787

RESUMEN

BACKGROUND: RevTet-On gene expression system was used to deliver the suicide gene tk to human breast cancer cell line MCF-7 and control the tk gene expression level. The animal model of human breast cancer on severe combined immune deficiency (SCID) mice was set up to explore the suicide gene therapy by the regulation of Tet-On. METHODS: Herpes simplex virus-thymidine kinase (HSVtk) gene was inserted into the plasmid pRevTRE and the recombinant retroviral vector pRevTRE/HSVtk was constructed. Using modified calcium phosphate co-precipitation method, two transfections, pRevTRE/HSVtk and pRevTet-On were performed for MCF-7 cell line and selected by hygromycin B and G418. MCF-7 cell line that stably expressed Tet-regulated tk gene was established. HSVtk gene expression in the MCF/TRE/tk/Tet-On cell line was under the control of Doxycycline (Dox). Cell viability was also determined by MTT assay, whereas HSVtk gene expression was analyzed by reverse transcription-PCR (RT-PCR). RESULTS: MCF/TRE/tk/Tet-On cell survival rate was decreased from 100% to less than 20% when ganciclovir (GCV) concentration was increased from 0 to 1000 microg/ml at 1 microg/ml of Dox after 72 hours of GCV administration. At 1 microg/ml of GCV concentration, the cell numbers decreased from 7 x 10(4) cells/ml to 2 x 10(4) cells/ml when Dox concentration was increased from 0 to 1500 ng/ml after 72 hours culture. In addition, bystander effects were generated in vitro when 10% - 25% of transduced MCF-7 cells were mixed in untransduced MCF-7 cells. On the other hand, the human breast cancer models in SCID mice were set up. The tk gene was expressed with the regulated character after MCF/TRE/tk/Tet-On cells were implanted into the female SCID mice 7 days after Dox induction followed by intraperitoneally administration of GCV for 23 days. Subcutaneous tumors in SCID mice that were implanted with MCF/TRE/tk/Tet-On cells shrank remarkably after Dox and GCV administration as compared with the control. CONCLUSION: The human breast tumor cells (MCF-7) expressing HSVtk gene can be eradicated by administration of GCV and induced with tetracycline or its derivative Dox in vitro and in vivo.


Asunto(s)
Neoplasias de la Mama/terapia , Genes Transgénicos Suicidas , Terapia Genética/métodos , Timidina Quinasa/genética , Animales , Efecto Espectador , Línea Celular Tumoral , Supervivencia Celular , Doxiciclina/farmacología , Ganciclovir/farmacología , Vectores Genéticos , Herpesviridae/genética , Humanos , Ratones , Ratones SCID , Retroviridae/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
19.
PLoS One ; 7(7): e40475, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22792345

RESUMEN

Our previous studies had shown that DAZAP2 was profoundly downregulated in bone marrow mononuclear cells from multiple myeloma patients. In this report, we analyzed epigenetic changes in multiple myeloma cell lines to understand the molecular mechanisms underlying the downregulation of DAZAP2. Four multiple myeloma cell lines, KM3, MM.1S, OPM-2 and ARH-77, were studied. The results of methylation specific PCR (MSP) showed that the promoter of DAZAP2 was methylated for KM3, MM.1S, OPM-2 and unmethylated for ARH-77. The DAZAP2 promoter region was amplified to obtain a series of different length sequences. All of the amplified sequences were inserted to luciferase reporter vector. The constructs were transfected into COS-7 cells and the luciferase activities were measured to search for the core region of DAZAP2 promoter. Two CpG islands were found in DAZAP2 promoter region. The results of luciferase assay showed that CpG island 1 displayed weak transcriptional activity, whereas CpG island 2 exhibited strong transcriptional activity (273 folds) compared to the control. The sequence that covered both CpG islands 1 and 2 showed higher activity (1,734 folds) compared to the control, suggesting that the two islands had synergistic effect on regulating DAZAP2 expression. We also found that M. Sss I methylase could inhibit the luciferase activity, whereas demethylation using 5-aza-2'-deoxycytidine treatment rescued the expression of DAZAP2 for multiple myeloma cell lines. These data revealed that methylation of DAZAP2 promoter was involved in downregulation of DAZAP2 in multiple myeloma cells.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Regiones Promotoras Genéticas , Proteínas de Unión al ARN/genética , Animales , Azacitidina/análogos & derivados , Azacitidina/farmacología , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Metilación de ADN , Metilasas de Modificación del ADN/antagonistas & inhibidores , Decitabina , Regulación hacia Abajo , Humanos , Mieloma Múltiple , Reacción en Cadena de la Polimerasa , Proteínas de Unión al ARN/metabolismo , Análisis de Secuencia de ADN , Transcripción Genética
20.
Sheng Wu Gong Cheng Xue Bao ; 21(3): 360-4, 2005 May.
Artículo en Zh | MEDLINE | ID: mdl-16108356

RESUMEN

In order to investigate the application of recombinant adeno-associated virus (rAAV) vector containing Tet regulation system and HSVtk gene in cancer gene therapy, pAAV/TRE/HSVtk/Tet-On was constructed and identified with PCR and restriction enzyme digestion. Packaging cells HEK293 were cotransfected with plasmids pAAV/TRE/HSVtk/Tet-On, pAAV-RC and pAAV-helper to produce infectious rAAV, and CsCl2 densitygradient centrifugation method was performed for purification and concentration of rAAV. The viruses were then transduced into MCF-7 cells. The results of dot blot hybridization indicate that the rAAV can transfer the target gene into MCF-7 cells. MTT assay showed that GCV could kill AAV-infected MCF-7 cells under the induction of Dox. The data demonstrated that rAAV containing Tet regulation system and HSVtk gene was successfully obtained, and could be used for further investigation of in vivo and in vitro experiments.


Asunto(s)
Dependovirus/genética , Genes Transgénicos Suicidas/genética , Simplexvirus/genética , Timidina Quinasa/genética , Línea Celular Tumoral , Dependovirus/metabolismo , Doxiciclina/farmacología , Ganciclovir/farmacología , Terapia Genética , Vectores Genéticos/genética , Humanos , Simplexvirus/enzimología , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA