Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Am J Hematol ; 97(5): 537-547, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35114022

RESUMEN

Few prospective studies have examined posttransplant chimeric antigen receptor (CAR) T cell infusion as candidates for front-line consolidation therapy for high-risk multiple myeloma (MM) patients. This single-arm exploratory clinical trial is the first to evaluate the safety and efficacy of sequential anti-CD19 and anti-BCMA CAR-T cell infusion, followed by lenalidomide maintenance after autologous stem cell transplantation (ASCT), in 10 high-risk newly diagnosed multiple myeloma (NDMM) patients. The treatment was generally well tolerated, with hematologic toxicities being the most common grade 3 or higher adverse events. All patients had cytokine release syndrome (CRS), which was grade 1 in 5 patients (50%) and grade 2 in 5 patients (50%). No neurotoxicity was observed after CAR-T cell infusion. The overall response rate was 100%, with the best response being 90% for a stringent complete response (sCR), and 10% for a complete response (CR). At a median follow-up of 42 (36-49) months, seven (70%) of 10 patients showed sustained minimal residual disease (MRD) negativity for more than 2 years. The median progression-free survival (PFS) and overall survival (OS) were not reached. Although the sample size was small and there was a lack of control in this single-arm study, the clinical benefits observed warrant ongoing randomized controlled trials.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Mieloma Múltiple , Humanos , Inmunoterapia Adoptiva/efectos adversos , Lenalidomida , Mieloma Múltiple/tratamiento farmacológico , Estudios Prospectivos , Trasplante Autólogo
2.
Pancreatology ; 18(7): 822-832, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30201439

RESUMEN

BACKGROUND: Cantharidin is an inhibitor of protein phosphatase 2 A (PP2A), and has been frequently used in clinical practice. In our previous study, we proved that cantharidin could arrest cell cycle in G2/M phase. Since cells at G2/M phase are sensitive to radiotherapy, in the present study, we investigated the radiotherapy-sesitization effect of cantharidin and the potential mechanisms involved. METHODS: Cell growth was determined by MTT assay. Cell cycle was evaluated by flow cytometry. DNA damage was visualized by phospho-Histone H2A.X staining. Expression of mRNA was tested by microarray assay and real-time PCR. Clinical information and RNA-Seq expression data were derived from The Cancer Genome Atlas (TCGA) pancreatic cancer cohort. Survival analysis was obtained by Kaplan-Meier estimates. RESULTS: Cantharidin strengthened the growth inhibition effect of irradiation. Cantharidin drove pancreatic cancer cells out of quiescent G0/G1 phase and arrested cell cycle in G2/M phase. As a result, cantharidin strengthened DNA damage which was induced by irradiation. Moreover, cantharidin repressed expressions of several genes participating in DNA damage repair, including UBE2T, RPA1, GTF2HH5, LIG1, POLD3, RMI2, XRCC1, PRKDC, FANC1, FAAP100, RAD50, RAD51D, RAD51B and DMC1, through JNK, ERK, PKC, p38 and/or NF-κB pathway dependent manners. Among these genes, worse overall survival for pancreatic cancer patients were associated with high mRNA expressions of POLD3, RMI2, PRKDC, FANC1, RAD50 and RAD51B, all of which could be down-regulated by cantharidin. CONCLUSION: Cantharidin can sensitize pancreatic cancer cells to radiotherapy. Multiple mechanisms, including cell cycle regulation, enhanced DNA damage, and inhibited DNA damage repair, may be involved.


Asunto(s)
Cantaridina/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Radioterapia , Ciclo Celular , División Celular/efectos de los fármacos , División Celular/efectos de la radiación , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Daño del ADN , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pancreáticas , Proteína Fosfatasa 2/antagonistas & inhibidores
3.
BMC Gastroenterol ; 18(1): 99, 2018 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-29954326

RESUMEN

BACKGROUND: The classifications and counts of white blood cells (WBCs) have been proved to be able to be used as prognostic markers in cancer cases. The present study investigated the potential values of the classifications and counts of WBC, including lymphocyte (LY), monocyte (MO), neutrophil (NE), eosinophil (EO), and basophil (BA) in the prognosis of resectable gastric cancers (GCs). METHODS: This retrospective study recruited 104 resectable GC cases which were pathologically confirmed. The patients were divided into two groups according to the median pre-treatment values. To evaluate the changes in WBC counts and classification after treatment, we introduced the concept of post/pre-treatment ratios (≤ 1 indicated count was not increased after therapy, while > 1 suggested increased count). RESULTS: Pre-treatment NE and total WBC counts were negatively correlated with overall survival (OS). Surgery significantly decreased the level of NE count, but increased the level of EO, whereas had no effect on the levels of LY, MO, BAor total WBC. Adjuvant chemotherapy significantly decreased the level of BA. Whole course of treatment (surgery combined with adjuvant chemotherapy) had no significant effect on the counts of LY, MO, NE, EO, BA or total WBC. Post/pre-treatment ratios of LY, MO NE, EO, BA and total WBC levels had no effects on OS. Univariate analysis indicated that AJCC stage (III) and higher level of pre-treatment total WBC count were prognostic factors affecting OS. Multivariate Cox regression analysis revealed that AJCC stage (III) and higher level of pre-treatment total WBC count were independent prognostic factors. CONCLUSIONS: Pre-treatment NE count and pre-treatment total WBC count may be potential prognostic factors for the prognostic evaluation of GCs.


Asunto(s)
Neoplasias Gástricas/clasificación , Neoplasias Gástricas/inmunología , Adulto , Anciano , Quimioterapia Adyuvante , Femenino , Humanos , Recuento de Leucocitos , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Neoplasias Gástricas/patología , Neoplasias Gástricas/cirugía , Análisis de Supervivencia
4.
Cancer Cell Int ; 17: 13, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28127258

RESUMEN

PURPOSE: Despite new developments in cancer therapy, chemotherapy and radiotherapy remain the cornerstone of breast cancer treatment. Therefore, finding ways to reduce the toxicity and increase sensitivity is particularly important. Tumor necrosis factor alpha (TNF-α) exerts multiple functions in cell proliferation, differentiation and apoptosis. In the present study, we investigated whether TNF-α could enhance the effect of chemotherapy and radiotherapy against breast cancer cells. METHODS: Cell growth was determined by MTT assay in vitro, and by using nude mouse tumor xenograft model in vivo. Cell cycle and apoptosis/necrosis were evaluated by flow cytometry. DNA damage was visualized by phospho-Histone H2A.X staining. mRNA expression was assessed by using real-time PCR. Protein expression was tested by Western blot assay. RESULTS: TNF-α strengthened the cytotoxicity of docetaxel, 5-FU and cisplatin against breast cancer cells both in vitro and in vivo. TNF-α activated NF-κB pathway and dependently up-regulated expressions of CyclinD1, CyclinD2, CyclinE, CDK2, CDK4 and CDK6, the key regulators participating in G1→S phase transition. As a result, TNF-α drove cells out of quiescent G0/G1 phase, entering vulnerable proliferating phases. Treatment of TNF-α brought more DNA damage after Cs137-irradiation and strengthened G2/M and S phase cell cycle arrest induced by docetaxel and cisplatin respectively. Moreover, the up-regulation of RIP3 (a necroptosis marker) by 5-FU, and the activation of RIP3 by TNF-α, synergistically triggered necroptosis (programmed necrosis). Knockdown of RIP3 attenuated the synergetic effect of TNF-α and 5-FU. CONCLUSION: TNF-α presented radiotherapy- and chemotherapy-sensitizing effects against breast cancer cells.

5.
Cancer Cell Int ; 15(1): 17, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25685069

RESUMEN

OBJECTIVE: Angiogenesis is a critical step of breast cancer metastasis. Oncogenic Ras promotes the remodeling of cancer microenviroment. Tumor-associated macrophages (TAMs) are a prominent inflammatory cell population emerging in the microenviroment and facilitating the angiogenesis and metastasis. In the present study, we tried to investigate the relationship between the expression of Ras and infiltration of TAM, both of which could further promote angiogenesis. METHODS: Expressions of Ras, CD68 and CD34 were assessed by immunohistochemistry. The infiltration of macrophages was evaluated by counting the number of CD68(+) cells. Vessel endothelial cells were defined as CD34(+) cells. Angiogenesis vascularity was defined by microvessel density (MVD) assay through counting the number of vessels per field counted in the area of highest vascular density. The Kaplan-Meier survival analysis was used to estimate the overall survival (OS). Macrophages were derived from monocytes in the presence of macrophage colony-stimulating-factor (MCSF). Breast cancer cells were treated with macrophage-conditioned medium (MCM) and tested the expressions of K-, H- and N-Ras by using realtime-PCR. RESULTS: Ras positive status was correlated with ER, PR and Her-2 positivity, larger tumour size and lymph node metastasis, as well as higher TNM stages. A higher number of CD68(+) cells was correlated with larger tumour size, higher TNM stages and Her-2 positivity. Both Ras positivity and infiltration of CD68(+) macrophages correlated with poor OS. The number of CD68(+) cells was positively correlated with the expression of Ras. Treatment with MCM did not up-regulate but repressed the expression of Ras. Both up-regulation of Ras and infiltration of TAMs correlated with increased MVD. CONCLUSION: Expression of Ras and infiltration of TAM were positively correlated, and both participated in angiogenesis. Elevated Ras could be responsible for the infiltration of TAM.

7.
Zhonghua Yi Xue Za Zhi ; 94(2): 86-9, 2014 Jan 14.
Artículo en Zh | MEDLINE | ID: mdl-24721345

RESUMEN

OBJECTIVE: To investigate the expression of protein phosphatase 2A catalytic subunit (PP2Ac) in pancreatic cancer and the regulation of this gene on JNK/c-Jun/AP-1 pathway and cell growth. METHODS: Expression of PP2Ac was determined by real-time PCR. Cell viability was tested by MTT.Expression and phosphorylation levels of proteins were detected by Western blotting. Cell cycle was assayed by flow cytometry. Transcription activity was measured by luciferase reporter gene assay. RESULTS: Suppressed PP2Ac expression was detected in pancreatic cancer tissues. PP2Ac expression in the pancreatic cancer cell lines was also at a low level.Overexpression of the two isoforms of PP2Ac, PP2Acα and PP2Acß, in pancreatic cancer cells repressed cell growth. Cell viability decreased (33.89 ± 2.05)% (t = 28.607, P < 0.001) and (16.66 ± 2.81)% (t = 10.257, P = 0.001) respectively 72 hours after transfection.Overexpression of PP2Acα and PP2Acß down-regulated the phosphorylation levels of JNK and c-Jun, and made the transcriptional activity of AP-1 decrease (47.18 ± 2.28)% (t = 11.230, P < 0.001) and (30.89 ± 8.09)% (t = 6.612, P = 0.003) respectively, indicating the down-regulation of PP2Ac up-regulated the activity of JNK/c-Jun/AP-1 pathway. Blocking the JNK pathway using a selective inhibitor, SP600125, induced G2/M cell cycle arrest and repressed cell proliferation. Cell viability decreased (31.38 ± 1.33)% (t = 40.930, P < 0.001) after treatment with JNK inhibitor for 72 hours. CONCLUSION: Suppressed expression of PP2Ac in pancreatic cancer facilitated cell growth through up-regulating the activity of JNK/c-Jun/AP-1 pathway.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Proteína Fosfatasa 2/metabolismo , Línea Celular Tumoral , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Neoplasias Pancreáticas
8.
Nucleic Acids Res ; 39(11): 4640-52, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21310710

RESUMEN

The 279-bp major breakpoint region (mbr) within the 3'-untranslated region (3'-UTR) of the BCL2 gene is a binding site of special AT-rich sequence binding protein 1 (SATB1) that is well known to participate in the long-range regulation of gene transcription. Our previous studies have revealed that the mbr could regulate BCL2 transcription over a 200-kb distance and this regulatory function was closely related to SATB1. This study is to explore the underlying mechanism and its relevance to cellular apoptosis. With chromosome conformation capture (3C) and chromatin immunoprecipitation (ChIP) assays we demonstrated that the mbr could physically interact with BCL2 promoter through SATB1-mediated chromatin looping, which was required for epigenetic modifications of the promoter, CREB accessibility and high expression of the BCL2 gene. During early apoptosis, SATB1 was a key regulator of BCL2 expression. Inhibition of SATB1 cleavage by treatment of cells with a caspase-6 inhibitor or overexpression of mutant SATB1 that was resistant to caspase-6, inhibited disassembly of the SATB1-mediated chromatin loop and restored the BCL2 mRNA level in Jurkat cells. These data revealed a novel mechanism of BCL2 regulation and mechanistically link SATB1-mediated long-range interaction with the regulation of a gene controlling apoptosis pathway for the first time.


Asunto(s)
Regiones no Traducidas 3' , Epigénesis Genética , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-bcl-2/genética , Apoptosis , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Cromosomas/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Humanos , Células Jurkat , Proteínas de Unión a la Región de Fijación a la Matriz/fisiología , Transcripción Genética , Factores de Transcripción p300-CBP/metabolismo
9.
Zhonghua Gan Zang Bing Za Zhi ; 21(6): 459-63, 2013 Jun.
Artículo en Zh | MEDLINE | ID: mdl-24034849

RESUMEN

OBJECTIVE: To generate a gene delivery plasmid carrying the dominant negative form of the protein phosphatase 2A catalytic subunit a (DN-PP2Aca) driven by a hepatocellular carcinoma (HCC) tissue-specific promoter and investigate its ability to inhibit growth of cultured hepatoma cells. METHODS: The gene delivery plasmid was constructed by PCR-amplifying DN-PP2Aca from wild-type PP2Aca using site-directed mutagenesis and then ligating the sequence-verified amplicon downstream of an alpha-fetoprotein enhancer and phosphoglycerate kinase promoter (AFpg) in the luciferase reporter vector pGL3-Basic. Following transfection into two AFP+ hepatoma cell lines (HepG2 and HepG3) and two AFP- hepatoma cell lines (SK-HEP-1 and L02), the transcriptional activity of the AFpg-driven DN-PP2Aca plasmid was tested using luciferase reporter gene assay and western blotting. The effect on cell growth was tested using MTT assay. Between group differences were assessed by t-test. RESULTS: The AFpg-driven DN-PP2Aca plasmid showed high transcriptional activity and protein expression in both HepG2 and Hep3B cells. At 72 h after transfection, the proliferation capacities were repressed by 42.65%+/-3.99% (P = 0.0002) and 39.87%+/-3.91% (P = 0.0002) in AFP+ HepG2 and Hep3B cells, respectively (vs. untransfected). In contrast, the plasmid was transcriptionally inactive in and had no effect on proliferation of AFP- cells. CONCLUSION: The AFpg-driven DN-PP2Aca plasmid exhibits selective cytotoxicity against AFP+ hepatoma cells, and may represent a useful gene therapy strategy to treat HCC.


Asunto(s)
Carcinoma Hepatocelular/genética , Vectores Genéticos , Neoplasias Hepáticas/genética , Proteína Fosfatasa 2/genética , Carcinoma Hepatocelular/metabolismo , Elementos de Facilitación Genéticos , Terapia Genética , Células Hep G2 , Humanos , Neoplasias Hepáticas/metabolismo , Mutación , Regiones Promotoras Genéticas , alfa-Fetoproteínas/genética
10.
J Gastrointest Oncol ; 14(3): 1525-1545, 2023 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-37435207

RESUMEN

Background: The incidence and mortality of pancreatic cancer are almost the same, and the 5-year survival rate is less than 10%. The high mortality of pancreatic cancer is related to chemo-radiotherapy. The present study aimed to establish a prognostic signature of pancreatic cancer based on chemo-radiotherapy resistant-related genes (CRRGs). Methods: In this study, we explored the radiation-resistant and chemotherapy-resistant pancreatic cancer cell lines by colony formation and a subcutaneous tumor model in nude mice. Next, we obtained CRRGs from radiation- and gemcitabine-resistant pancreatic cancer cell lines in the Gene Expression Omnibus (GEO) database. Based on univariate Cox and least absolute shrinkage and selection operator (LASSO) Cox regression analyses, a prognostic model of the pancreatic adenocarcinoma (PAAD) cohort in The Cancer Genome Atlas (TCGA) database (N=177) was established and verified using the GEO cohort data set (N=112). Finally, the functions of candidate target genes were verified by a methyl thiazolyl tetrazolium (MTT) assay, a colony formation assay, and a subcutaneous tumor model in nude mice. Results: Through the in vitro and in vivo experiments, we found that radiotherapy- and chemotherapy-resistant pancreatic cancer cells were cross-resistant to chemotherapy and radiotherapy. We constructed a risk model consisting of nine CRRGs (SNAP25, GPR87, DLL1, LAD1, WASF3, ARHGAP29, ZBED2, GAD1, and JAG1) by using public databases. According to the Kaplan-Meier curve analysis, the survival of the high-risk group was worse than that of the low-risk group. We then used nomograms to predict the 1/3/5-year overall survival (OS) in pancreatic cancer patients. We chose JAG1 as a candidate target since it has been proven to be involved in the stemness maintenance of cancer cells, and found that JAG1 silencing inhibited the proliferation and chemo-radiotherapy tolerance of pancreatic cancer cells. Conclusions: This study established and validated a prognostic signature of pancreatic cancer using nine CRRGs. The in vitro and in vivo experiments showed that JAG1 could promote the proliferation and chemoradiotherapy tolerance of pancreatic cancer cell lines. These findings may offer new insights into the role of CRRGs in pancreatic cancer and provide novel prognostic biomarkers for the treatment of pancreatic cancer.

11.
ACS Appl Mater Interfaces ; 15(15): 18808-18818, 2023 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-37036119

RESUMEN

A proton (H+) and zinc ion (Zn2+) co-insertion model is put forward in this study to elucidate the capacity origin of an aqueous zinc ion battery (ZIB) based on a heavily loaded (∼15 mg cm-2) cathode, which consists of Na2V6O16·3H2O (NVO) embedded particularly in the macropores of activated carbon cloth (ACC), coupled with a highly stable Zn/In anode. The confinement effect of these porous channels not only prevents the detachment of NVO from ACC but also well mitigates its volume change resulting from H+ and Zn2+ co-intercalation, which collectively render the stability of NVO/ACC markedly enhanced. Moreover, the bicontinuous structure of NVO/ACC, as a result of the self-interlacing of intrapore NVO, which is first engineered into the nanobelts, and their interlocking with the carbon fibers of ACC, simultaneously giving rise to a solid and a holey framework, is favorable to the electron and ion transport throughout the entire electrode. The synergistic effect of such facile charge transfer kinetics and the high packing density of NVO in the cathode endows ZIBs with not only a good rate performance but also an exceptional areal capacity amounting to 4.6 mAh cm-2, far surpassing those reported for additional vanadium-based counterparts reported in the literature.

12.
BMC Cancer ; 12: 547, 2012 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-23173703

RESUMEN

BACKGROUND: Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related deaths worldwide. Current therapies are insufficient, making HCC an intractable disease. Our previous studies confirmed that inhibition of protein phosphatase 2A (PP2A) may provide a promising therapeutic strategy for cancer. Unfortunately, constitutive expression of PP2A in normal tissues limits the application of PP2A inhibition. Thus, a HCC-specific gene delivery system should be developed. The α-fetoprotein (AFP) promoter is commonly used in HCC-specific gene therapy strategies; however, the utility of this approach is limited due to the weak activity of the AFP promoter. It has been shown that linking the AFP enhancer with the promoter of the non-tissue-specific, human housekeeping phosphoglycerate kinase (pgk) gene can generate a strong and HCC-selective promoter. METHODS: We constructed a HCC-specific gene therapy system to target PP2A using the AFP enhancer/pgk promoter, and evaluated the efficiency and specificity of this system both in vitro and in vivo. RESULTS: AFP enhancer/pgk promoter-driven expression of the dominant negative form of the PP2A catalytic subunit α (DN-PP2Acα) exerted cytotoxic effects against an AFP-positive human hepatoma cell lines (HepG2 and Hep3B), but did not affect AFP-negative human hepatoma cells (SK-HEP-1) or normal human liver cells (L-02). Moreover, AFP enhancer/pgk promoter driven expression of DN-PP2Acα inhibited the growth of AFP-positive HepG2 tumors in nude mice bearing solid tumor xenografts, but did not affect AFP-negative SK-HEP-1 tumors. CONCLUSIONS: The novel approach of AFP enhancer/pgk promoter-driven expression of DN-PP2Acα may provide a useful cancer gene therapy strategy to selectively target HCC.


Asunto(s)
Carcinoma Hepatocelular/terapia , Terapia Genética/métodos , Neoplasias Hepáticas Experimentales/terapia , Proteínas de Neoplasias/antagonistas & inhibidores , Fosfoglicerato Quinasa/genética , Proteína Fosfatasa 2/antagonistas & inhibidores , alfa-Fetoproteínas/genética , Animales , Apoptosis/fisiología , Cantaridina/farmacología , Carcinoma Hepatocelular/enzimología , Ciclo Celular/fisiología , Línea Celular Tumoral , Elementos de Facilitación Genéticos , Inhibidores Enzimáticos/farmacología , Humanos , Neoplasias Hepáticas Experimentales/enzimología , Ratones , Ratones Desnudos , Proteínas de Neoplasias/fisiología , Regiones Promotoras Genéticas , Proteína Fosfatasa 2/fisiología
13.
Leuk Lymphoma ; 63(11): 2636-2644, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35731036

RESUMEN

Leukapheresis is an effective adjuvant therapy for leukemia patients with hyperleukocytosis, but few studies have reported recent data with modern modalities and comparisons among different leukemia types. We conducted a retrospective study on leukapheresis among 420 patients with AML, ALL and CML in four local centers. WBC counts decreased significantly (p < 0.001) postleukapheresis in all three cohorts. Clearance efficiency was higher in acute leukemia patients than CML patients (p < 0.01). Concomitant leukocytoreduction drugs improved WBC reduction only in AML patients (p < 0.05). Leukocyte, hemoglobin and platelet levels preleukapheresis might affect the clearance efficiency in AML and/or ALL patients. Hematological toxicities were the major concerns, but most of them were mild, and only 11 patients died of all causes within one week postleukapheresis. In conclusion, leukapheresis can safely reduce the leukemic burden, especially for patients with acute leukemias.


Asunto(s)
Leucaféresis , Leucemia Mieloide Aguda , Humanos , Estudios Retrospectivos , Leucocitosis/terapia , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/terapia , Leucemia Mieloide Aguda/complicaciones , Recuento de Leucocitos , Enfermedad Aguda
14.
J Cell Biochem ; 110(5): 1208-18, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20564216

RESUMEN

BCL2, originally identified as a proto-oncogene in B-cell lymphoma, is a key regulator of apoptosis. Although it is more than 200 kb in length, at least 70% of the t(14;18) translocation in follicular lymphomas occurs at the BCL2 major breakpoint region (mbr), located in the 3'-untranslated region (3'-UTR). We have previously found that the mbr is a regulatory element which positively regulates BCL2 expression and this regulatory function was closely associated with SATB1, which binds to a 37 bp mbr (37 mbr) in the 3'-end of the mbr directly. However, the precise molecular mechanisms by which the mbr regulates gene expression are not fully understood. In this study, we purified Poly(ADP-ribose) polymerase-1 (PARP-1) from the DNA-protein complexes formed by 37 mbr in Jurkat cells and demonstrated that PARP-1 participates in the 37 mbr-protein complex's formation in vitro and in vivo. Functional analysis showed that overexpression of PARP-1 decreases 37 mbr regulatory function and BCL2 expression. Conversely, knockdown of PARP-1 with RNAi increases BCL2 expression. Taken together, the present findings indicate that PARP-1 is a component of BCL2 37 mbr-protein complexes, and PARP-1 is involved in the regulation of BCL2 expression. These findings are helpful in understanding the regulatory mechanisms of BCL2 expression.


Asunto(s)
Puntos de Rotura del Cromosoma , Regulación Neoplásica de la Expresión Génica , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Regiones no Traducidas 3'/genética , Western Blotting , Línea Celular , Línea Celular Tumoral , Humanos , Células Jurkat , Poli(ADP-Ribosa) Polimerasas/genética , Unión Proteica , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-bcl-2/genética , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
15.
Oncol Lett ; 18(1): 571-580, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31289529

RESUMEN

Colorectal cancer (CRC) represents the third most common malignancy worldwide. The aim of the present study was to investigate the predictive values of platelet-associated indicators, including platelet count (PLT), plateletcrit (PCT), mean platelet volume (MPV) and platelet distribution width (PDW) in patients with resectable CRC. The current retrospective study included 153 patients who were pathologically diagnosed with resectable CRC. The patients were divided into two groups according to the median value of PLT, PCT, MPV or PDW. To evaluate the changes in PLT, PCT, MPV and PDW following resection and adjuvant chemotherapy, the concept of post-/pre-treatment PLT, PCT, MPV and PDW ratios was introduced, where <1 indicated decreased PLT, PCT, MPV and PDW values after treatment, and where ≥1 suggested stable or increased values. It was revealed that a low MPV prior to treatment correlated with a higher tumor stage. Surgery significantly decreased MPV, but had no impact on PLT, PCT or PDW. Adjuvant chemotherapy significantly decreased PLT and PCT, increased MPV and had no effect on PDW. After the whole course of treatment (surgery combined with adjuvant chemotherapy), PLT, PCT and PDW were significantly decreased. Kaplan-Meier plots illustrated that patients with a post-/pre-treatment MPV ratio <1 had poorer overall survival (OS), whereas the post-/pre-treatment ratios for PLT, PCT and PDW did not correlate with patient outcome. Multivariate Cox regression analysis revealed that sex, tumor size and the post-/pre-treatment MPV ratio were prognostic factors for OS. Therefore, the present results may suggest MPV as a potential prognostic factor in resectable CRC.

16.
Technol Cancer Res Treat ; 18: 1533033819837261, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30871415

RESUMEN

BACKGROUND: Lung cancer is the leading cause of cancer death. Platelet-related indictors, including platelet count, plateletcrit, mean platelet volume, and platelet distribution width, not only associate with morphology and functions of platelet but also correlate with tumor development and metastasis. In the present study, we investigated the values of platelet-related indictors in the prognosis evaluation of resectable lung cancers. METHODS: In total, 101 patients with resectable lung cancer were recruited in this study. Patients were divided into 2 groups according to the median pretreatment values. To evaluate the individual value changes after treatment, we introduced the concept of post-/pretreatment ratio (≤1 indicated value was not increased after treatment, while >1 suggested increased value). RESULTS: The high pretreatment platelet count level was correlated with larger tumor size. High pretreatment plateletcrit level was associated with more lymph nodes metastasis. Patients with high pretreatment plateletcrit level had worse overall survival, whereas pretreatment platelet count, mean platelet volume, and platelet distribution width levels were not correlated with outcomes. Surgery had no impact on the values of platelet count, plateletcrit, mean platelet volume, or platelet distribution width. Adjuvant chemotherapy significantly decreased the values of platelet count and plateletcrit, whereas it had no effect on the values of mean platelet volume or platelet distribution width. Whole course of treatment (surgery combined with adjuvant chemotherapy) significantly decreased the values of platelet count and platelet distribution width, whereas it had no effect on the values of plateletcrit or mean platelet volume. Post-/pretreatment platelet count, plateletcrit, mean platelet volume, and platelet distribution width ratios were not correlated with outcomes. Univariate analyses demonstrated that American Joint Committee on Cancer stage and pretreatment plateletcrit level were significant risk factors for prognosis. Cox regression analysis revealed that no factor independently associated with worse survival. CONCLUSION: Pretreatment plateletcrit level could be a potential prognostic factor in resectable lung cancers.


Asunto(s)
Plaquetas/patología , Carcinoma de Pulmón de Células no Pequeñas/secundario , Neoplasias Pulmonares/patología , Carcinoma Pulmonar de Células Pequeñas/secundario , Plaquetas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/cirugía , Femenino , Estudios de Seguimiento , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/cirugía , Metástasis Linfática , Masculino , Volúmen Plaquetario Medio , Persona de Mediana Edad , Invasividad Neoplásica , Pronóstico , Estudios Retrospectivos , Carcinoma Pulmonar de Células Pequeñas/metabolismo , Carcinoma Pulmonar de Células Pequeñas/cirugía , Tasa de Supervivencia
17.
Oncol Lett ; 17(2): 2244-2256, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30675290

RESUMEN

Lung cancer is one of the leading causes of cancer-associated mortality. C-reactive protein (CRP), albumin (ALB), globulin (GLB), lactate dehydrogenase (LDH), neutrophil-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) have been identified as general parameters for systemic inflammatory response (SIR). Furthermore, these parameters are also associated with tumor development and metastasis. The present study aimed to investigate the predictive values of these SIR parameters in patients with resectable lung cancer. In total, 101 patients with resectable lung cancer were recruited in the present study. The patients were divided into two groups according to the median value of pre-treatment CRP, ALB, GLB, LDH, NLR or PLR values. The post-/pre-treatment ratios were defined as the ratio of pre-treatment blood parameter values and the corresponding values obtained following therapy. A ratio of ≤1.1 indicated that the values were not increased, while a ratio of >1.1 suggested that the values were increased following treatment. Patients with lower pre-treatment ALB levels had poorer overall survival (OS) rates, whereas GLB, LDH, CRP, NLR or PLR levels were not associated with outcomes. Whole course treatment (surgery combined with adjuvant chemotherapy) significantly increased the value of ALB, but decreased the value of NLR, whereas it had no effect on the values of LDH, CRP or PLR. Post-/pre-treatment LDH and PLR were associated with outcomes. Post-/pre-treatment ALB, GLB, CRP and NLR were not associated with outcomes. Multivariate analysis revealed that a low pre-treatment ALB level and increased post-/pre-treatment PLR were independent risk factors affecting OS. The receiver operating characteristic curve analysis demonstrated that an ALB value of 47.850 g/l was considered to be the optimal cut-off value for prognosis; the sensitivity was 28.8% and specificity was 95.9%. It was suggested that the pre-treatment ALB and post-/pre-treatment PLR may be potential prognostic factors in resectable lung cancer.

18.
Mol Med Rep ; 19(3): 2330-2340, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30664202

RESUMEN

The count and classification of white blood cells (WBCs) may be used as prognostic markers in certain types of cancer. The present study investigated the prognostic potential of the counts of WBCs, including lymphocytes (LYs), monocytes (MOs), neutrophils (NEs), eosinophils (EOs) and basophils (BAs), in the prognosis of resectable colorectal cancer. The present study recruited 153 resectable colorectal cancer cases retrospectively, which were pathologically confirmed. All patients were divided into two groups, according to the median value of LY (low LY, ≤1.632x109/l or high LY, >1.632x109/l), MO (low MO, ≤0.330x109/l or high MO, >0.330x109/l), NE (low NE, ≤3.600x109/l or high NE, >3.600x109/l), EO (low EO, ≤0.085x109/l or high EO, >0.085x109/l), BA (low BA, ≤0.010x109/l or high BA, >0.010x109/l), or WBC (low WBC, ≤5.780x109/l or high WBC, >5.780x109/l). To evaluate the alterations in WBC counts following surgery and adjuvant chemotherapy; all samples received oxiplatin and capecitabine (XELOX) for 6­8 cycles or 5­fluorouracil, leucovorin and oxaliplatin (mFOLFOX6) for 10­12 cycles. XELOX included oxaliplatin administered intravenously at a dose of 130 mg/m2 on day 1 and 850­1,250 mg/m2 capecitabine twice daily for days 1­14, repeated every 3 weeks. mFOLFOX6 included oxaliplatin administered intravenously at a dose of 85 mg/m2, 400 mg/m2 leucovorin and 400 mg/m2 5­FU on day 1 followed by 1,200 mg/m2/days continuous infusion for 2 days (in total, 2,400 mg/m2 over 46­48 h), repeated every 2 weeks. The present study investigated the post/pre­treatment of LY, MO, NE, EO, BA and WBC ratios (≤1 indicated that LY, MO, NE, EO, BA and WBC counts were not increased following therapy; whereas, >1 suggested increased counts). Kaplan­Meier curves were constructed to demonstrate overall survival (OS). A multivariate and univariate logistic regression analyses model was employed to identify the independent risk factors. Low pre­treatment BA counts were associated with larger tumor size (>5 cm); pre­treatment BA levels were positively associated with OS. Surgery significantly decreased the count of BAs and increased the count of EOs; whereas, no effect was observed on LYs, MOs, NEs or WBCs. Adjuvant chemotherapy markedly decreased the counts of LY, NE and WBC; whereas, no notable effects on MOs, EOs or BAs were observed. Whole course treatment (surgery combined with adjuvant chemotherapy) significantly decreased the values of LY, NE and WBC; however, increased the value of EO; no effects on the MO or BA counts were observed. An increased post­/pre­treatment NE ratio suggested poorer prognosis. Multivariate Cox regression analysis revealed that sex, tumor size, pre­treatment BA count and the post­/pre­treatment NE ratio were independent prognostic factors affecting OS. The results of the present study suggested that the pre­treatment BA count and post­/pre­treatment NE ratio may be potential prognostic factors for resectable colorectal cancer.


Asunto(s)
Neoplasias Colorrectales/sangre , Neoplasias Colorrectales/tratamiento farmacológico , Recuento de Leucocitos , Pronóstico , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Basófilos/efectos de los fármacos , Capecitabina , Quimioterapia Adyuvante , Neoplasias Colorrectales/patología , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Eosinófilos/efectos de los fármacos , Femenino , Fluorouracilo/administración & dosificación , Fluorouracilo/análogos & derivados , Humanos , Leucovorina/administración & dosificación , Linfocitos/efectos de los fármacos , Masculino , Persona de Mediana Edad , Neutrófilos/efectos de los fármacos , Compuestos Organoplatinos/administración & dosificación , Oxaloacetatos
19.
Oncogenesis ; 7(11): 94, 2018 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-30478299

RESUMEN

Cantharidin, one of the active components of mylabris, is believed to have antitumor activity. Cantharidin selectively inhibits protein phosphatase 2A (PP2A), which can repress multiple oncogenic kinases (ERK, JNK, PKC, and NF-κB). Researches in vitro have shown that cantharidin suppresses cell viability and metastasis in pancreatic cancer cells. This study aims to investigate the effects of cantharidin on pancreatic cancer xenografts in vivo. Xenograft models were established using cells stably expressing luciferase. Xenograft growth was evaluated by living imaging. Gene expression was determined using a microarray, real-time PCR, a RayBiotech antibody array, and the Milliplex assay. Surprisingly, cantharidin significantly accelerated xenograft growth. Living imaging showed a rapid distribution of D-luciferin in cantharidin-treated xenografts, suggesting a rich blood supply. Immunohistochemistry confirmed increased angiogenesis. Microarray and antibody array identified upregulated proangiogenic and downregulated antiangiogenic factors. The Milliplex assay suggested elevated secretion of IL-6, IL-8, TNF-α, and VEGF. Inhibitors of ERK, JNK, PKC, and NF-κB pathway attenuated the cantharidin-induced changes to proangiogenic gene expression. PKC pathway-inhibiting tamoxifen or antiangiogenic therapeutics, including Ginsenoside Rg3, bevacizumab, Apatinib, and Endostar, antagonized the proangiogenic effect of cantharidin or its derivatives. These regimens presented remarkable additive antitumor effects in vivo. Although cantharidin presents antitumor effects in vitro and has been applied in clinical practice, we revealed an unfavorable proangiogenic side effect. We recommend that the clinical application of cantharidin should be performed on the premise of antivascularization therapy.

20.
Int J Oncol ; 52(1): 127-138, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29115601

RESUMEN

Anti-angiogenic therapy has been successfully applied to treat colorectal cancer (CRC). Ginsenoside Rg3, derived from the Chinese herb ginseng, has anti-vascularization effects and can inhibit tumor growth and metastasis, and can sensitize cancer cells to chemotherapy. Therefore, in the present study, we investigated whether Rg3 could be appropriate for CRC treatment. Growth of CRC cells was assessed by an MTT (methyl thiazolyl tetrazolium) assay in vitro and using orthotopic xenograft models in vivo. mRNA expression was evaluated using real-time PCR. Protein levels were tested by western blotting, flow cytometry and immunohistochemistry. Migration was determined using a wound-healing assay. Stemness was further confirmed using a plate clone formation assay. We found that Rg3 repressed the growth and stemness of CRC cells both in vitro and in vivo. Rg3 also impaired the migration of CRC cells in vitro. Rg3 downregulated the expressions of angiogenesis-related genes, and repressed the vascularization of CRC xenografts. In addition, Rg3 strengthened the cytotoxicity of 5-Fluorouracil and oxaliplatin against orthotopic xenografts in vivo. Moreover, Rg3 downregulated the expressions of B7-H1 and B7-H3, high expressions of which were associated with reduced overall survival (OS) of CRC patients. Hence, Rg3 not only repressed the growth and stemness of CRC cells, but could also remodel the tumor microenvironment through repressing angiogenesis and promoting antitumor immunity. Therefore, Rg3 could be a novel therapeutic for the CRC treatment.


Asunto(s)
Neoplasias Colorrectales/tratamiento farmacológico , Ginsenósidos/farmacología , Células Madre Neoplásicas/efectos de los fármacos , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Neoplasias Colorrectales/irrigación sanguínea , Neoplasias Colorrectales/patología , Progresión de la Enfermedad , Sinergismo Farmacológico , Femenino , Fluorouracilo/administración & dosificación , Fluorouracilo/farmacología , Ginsenósidos/administración & dosificación , Células HCT116 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Compuestos Organoplatinos/administración & dosificación , Compuestos Organoplatinos/farmacología , Oxaliplatino , Distribución Aleatoria , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA