Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Int J Cancer ; 139(1): 171-6, 2016 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26891175

RESUMEN

Anecdote clinical observations hint that non-small cell lung cancer (NSCLC) with exon-20 insertions might respond poorly to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs), contrasting to those with classic mutations. Lack of patient-derived experimental models has been a major hurdle for the discovery of new treatment for the diseases. We established two NSCLC-PDXs harboring two different exon-20 insertions, LU0387-adenocarcinoma (ADC) with a nine-base insertion at 2319 (H773-V774insNPH) and LU3075-squamous cell carcinoma (SCC) with a nine-base insertion at 2316 (P772-H773insDNP). Both insertions immediately follow the regulatory C-helix of the kinase domain. Contrary to the generally good responses to EGFR inhibitors observed in PDXs with classic mutations, both exon-20 insertions are largely resistant to cetuximab and TKIs in vivo, suggesting fundamental difference from the classic EGFR mutations, consistent with the poor response rate to TKI seen in anecdotal clinic reports. It is worth noting that although responses are generally poor, they differ between the two exon-20 mutants depending on the type of TKI. In vitro drug sensitivity assays using established primary cell lines from our two PDXs largely confirmed the in vivo data. Our data from patient-derived experimental models confirmed that exon-20 insertions in domain immediately following the C-helix confer poor response to all known EGFR inhibitors, and suggested that these models can be utilized to facilitate the discovery of new therapies targeting NSCLC harboring exon-20 insertions.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Células Escamosas/tratamiento farmacológico , Receptores ErbB/genética , Adenocarcinoma/genética , Adenocarcinoma/patología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Receptores ErbB/antagonistas & inhibidores , Exones/genética , Humanos , Mutación , Inhibidores de Proteínas Quinasas , Quinazolinas/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
2.
PLoS One ; 19(8): e0309394, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39172988

RESUMEN

GDF15 (growth differentiation factor 15), also known as macrophage inhibitory cytokine 1 (MIC-1), is a circulating protein involved in the regulation of energy balance and weight control. Elevated levels of GDF15 have been associated with cachexia and reduced survival rates in cancer patients. Through the activation of the GFRAL (GDNF-family receptor α-like)-RET (Rearranged during Transfection) signaling pathway, GDF15 can induce weight loss, making it a potential target for treating cachexia. Currently, there are no approved antibody drugs specifically targeting GDF15 for cancer cachexia treatment. However, efforts have been made to develop antibody-based therapeutics against this emerging target. In this study, we generated a monoclonal antibody KY-NAb-GDF15 against GDF15 that effectively blocks downstream signaling mediated by GFRAL upon stimulation by GDF15. This antibody demonstrates robust neutralizing activity and exhibits high binding specificity. Importantly, our findings indicate that this antibody holds promise in alleviating cancer-induced cachexia and mitigating chemotherapy-induced weight loss, thereby offering significant therapeutic potential for managing cancer cachexia.


Asunto(s)
Anticuerpos Neutralizantes , Caquexia , Factor 15 de Diferenciación de Crecimiento , Neoplasias , Caquexia/tratamiento farmacológico , Caquexia/inmunología , Anticuerpos Neutralizantes/uso terapéutico , Humanos , Neoplasias/complicaciones , Neoplasias/inmunología , Animales , Ratones , Línea Celular Tumoral , Transducción de Señal/efectos de los fármacos , Anticuerpos Monoclonales/uso terapéutico , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo
3.
Sci Rep ; 14(1): 9032, 2024 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-38641704

RESUMEN

CSF1R is a receptor tyrosine kinase responsible for the growth/survival/polarization of macrophages and overexpressed in some AML patients. We hypothesized that a novel multi-kinase inhibitor (TKi), narazaciclib (HX301/ON123300), with high potency against CSF1R (IC50 ~ 0.285 nM), would have anti-AML effects. We tested this by confirming HX301's high potency against CSF1R (IC50 ~ 0.285 nM), as well as other kinases, e.g. FLT3 (IC50 of ~ 19.77 nM) and CDK6 (0.53 nM). An in vitro proliferation assay showed that narazaciclib has a high growth inhibitory effect in cell cultures where CSF1R or mutant FLT3-ITD variants that may be proliferation drivers, including primary macrophages (IC50 of 72.5 nM) and a subset of AML lines (IC50 < 1.5 µM). In vivo pharmacology modeling of narazaciclib using five AML xenografts resulted in: inhibition of MV4-11 (FLT3-ITD) subcutaneous tumor growth and complete suppression of AM7577-PDX (FLT3-ITD/CSF1Rmed) systemic growth, likely due to the suppression of FLT3-ITD activity; complete suppression of AM8096-PDX (CSF1Rhi/wild-type FLT3) growth, likely due to the inhibition of CSF1R ("a putative driver"); and nonresponse of both AM5512-PDX and AM7407-PDX (wild-type FLT3/CSF1Rlo). Significant leukemia load reductions in bone marrow, where disease originated, were also achieved in both responders (AM7577/AM8096), implicating that HX301 might be a potentially more effective therapy than those only affecting peripheral leukemic cells. Altogether, narazaciclib can potentially be a candidate treatment for a subset of AML with CSF1Rhi and/or mutant FLT3-ITD variants, particularly second generation FLT3 inhibitor resistant variants.


Asunto(s)
Antineoplásicos , Leucemia Mieloide Aguda , Inhibidores de Proteínas Quinasas , Humanos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/metabolismo , Tirosina Quinasa 3 Similar a fms/antagonistas & inhibidores , Tirosina Quinasa 3 Similar a fms/metabolismo , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/patología , Mutación , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Tirosina Quinasas Receptoras , Receptores del Factor Estimulante de Colonias/antagonistas & inhibidores , Receptores del Factor Estimulante de Colonias/metabolismo , Piridonas/farmacología , Pirimidinas/farmacología
4.
ACS Chem Biol ; 2024 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-39283696

RESUMEN

Mutated KRAS proteins are frequently expressed in some of the most lethal human cancers and thus have been a target of intensive drug discovery efforts for decades. Lately, KRAS(G12C) switch-II pocket (SII-P)-targeting covalent small molecule inhibitors have finally reached clinical practice. Sotorasib (AMG-510) was the first FDA-approved covalent inhibitor to treat KRAS(G12C)-positive nonsmall cell lung cancer (NSCLC), followed soon by adagrasib (MRTX849). Both drugs target the GDP-bound state of KRAS(G12C), exploiting the strong nucleophilicity of acquired cysteine. Here, we evaluate the similarities and differences between sotorasib and adagrasib in their RAS SII-P binding by applying biochemical, cellular, and computational methods. Exact knowledge of SII-P engagement can enable targeting this site by reversible inhibitors for KRAS mutants beyond G12C. We show that adagrasib is strictly KRAS- but not KRAS(G12C)-specific due to its strong and unreplaceable interaction with H95. Unlike adagrasib, sotorasib is less dependent on H95 for its binding, making it a RAS isoform-agnostic compound, having a similar functionality also with NRAS and HRAS G12C mutants. Our results emphasize the accessibility of SII-P beyond oncogenic G12C and aid in understanding the molecular mechanism behind the clinically observed drug resistance, associated especially with secondary mutations on KRAS H95 and Y96.

5.
Mol Pharmacol ; 82(5): 938-47, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22899868

RESUMEN

Interleukin-2-inducible T-cell kinase (Itk) is a member of the Btk (Bruton's tyrosine kinase) family of tyrosine kinases. Itk plays an important role in normal T-cell functions and in the pathophysiology of both autoimmune diseases and T-cell malignancies. Here, we describe the initial characterization of a selective inhibitor, 7-benzyl-1-(3-(piperidin-1-yl)propyl)-2-(4-(pyridin-4-yl)phenyl)-1H-imidazo[4,5-g]quinoxalin-6(5H)-one (CTA056), that was developed through screening a 9600-compound combinatorial solution phase library, followed by molecular modeling, and extensive structure-activity relationship studies. CTA056 exhibits the highest inhibitory effects toward Itk, followed by Btk and endothelial and epithelial tyrosine kinase. Among the 41 cancer cell lines analyzed, CTA056 selectively targets acute lymphoblastic T-cell leukemia and cutaneous T-cell lymphoma. Normal T cells are minimally affected. Incubation of Jurkat and MOLT-4 cells with CTA056 resulted in the inhibition of the phosphorylation of Itk and its effectors including PLC-γ, Akt, and extracellular signal-regulated kinase, as well as the decreased secretion of targeted genes such as interleukin-2 and interferon-γ. Jurkat cells also underwent apoptosis in a dose-dependent manner when incubated with CTA056. The potent apoptosis-inducing potential of CTA056 is reflected by the significant modulation of microRNAs involved in survival pathways and oncogenesis. The in vitro cytotoxic effect on malignant T cells is further validated in a xenograft model. The selective expression and activation of Itk in malignant T cells, as well as the specificity of CTA056 for Itk, make this molecule a potential therapeutic agent for the treatment of T-cell leukemia and lymphoma.


Asunto(s)
Antineoplásicos/química , Bencimidazoles/química , Interleucina-2/antagonistas & inhibidores , MicroARNs/metabolismo , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Quinazolinas/química , Linfocitos T/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Bencimidazoles/síntesis química , Bencimidazoles/farmacología , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Interferón gamma/antagonistas & inhibidores , Interferón gamma/metabolismo , Interleucina-2/metabolismo , Linfoma Cutáneo de Células T , Ratones , Ratones Desnudos , Modelos Moleculares , Trasplante de Neoplasias , Fosforilación , Leucemia-Linfoma Linfoblástico de Células T Precursoras , Proteínas Tirosina Quinasas/metabolismo , Quinazolinas/síntesis química , Quinazolinas/farmacología , Linfocitos T/enzimología , Linfocitos T/patología , Trasplante Heterólogo , Regulación hacia Arriba
6.
J Cell Biochem ; 103(3): 730-9, 2008 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17551969

RESUMEN

Pleiomorphic adenoma gene (PLAG) family proteins are oncogenes involved in various malignancies including lipoblastomas, hepatoblastomas, and acute myeloid leukemia. Overexpression of PLAGL2 induces cell transformation and proliferation, but little is known about how its activities are regulated. We previously showed that transcriptional activity of PLAGL2 is negatively regulated by sumoylation. Here we report that Tip60 modulates PLAGL2 functions through acetylation. Tip60 associates with PLAGL2 through its zinc finger domain and acetylates PLAGL2. Wild-type but not the histone acetyltransferase (HAT)-minus mutant form of Tip60 enhances PLAGL2-mediated transactivation. In addition, coexpression of Tip60 and PLAGL2 completely abolishes the sumoylation of PLAGL2. Both Tip60 and DN-Ubc9 increase transactivation activity of wild-type but not the sumoylation deficient form of PLAGL2 (K250, 269, 356R), indicating that Tip60 acetylates PLAGL2 and abolishes the sumoylation of PLAGL2 possibly through modification of the same lysine residues (K250, 269, 356) within PLAGL2. Tip60 effects vary between different PLAGL2 target gene promoters, suggesting that Tip60 is a novel promoter-specific coactivator of PLAGL2. This is the first demonstration that Tip60 can function as a sumoylation inhibitor in part through its intrinsic acetyltransferase activity to regulate specific gene expression.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Histona Acetiltransferasas/metabolismo , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/metabolismo , Activación Transcripcional , Acetilación , Animales , Línea Celular , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Embrión de Mamíferos , Histona Acetiltransferasas/genética , Humanos , Riñón , Luciferasas/genética , Lisina/química , Lisina Acetiltransferasa 5 , Regiones Promotoras Genéticas/genética , Procesamiento Proteico-Postraduccional/genética , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Proteína SUMO-1/genética , Proteína SUMO-1/metabolismo , Factores de Transcripción/química , Factores de Transcripción/genética
7.
Biochem Biophys Res Commun ; 364(2): 344-50, 2007 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-17950244

RESUMEN

PLAGL2 (Pleomorphic Adenoma Gene Like 2) is an oncoprotein involved in various malignancies including lipoblastomas, hepatoblastomas, and acute myeloid leukemia. Although PLAGL2 is known to mainly act as a transcription factor, other functions which may contribute to its oncogenic potential are not clear. Pirh2 (P53 induced RING-H2 protein) is a p53 inducible E3 ligase involved in the ubiquitination of p53, while the mechanisms to regulate its activities are largely unknown. In this study, we show for the first time that Pirh2 forms dimers through its N- and C-terminus in cells and Pirh2 dimers interact with PLAGL2. The interaction between PLAGL2 and Pirh2 dimers prevents proteasomal degradation of Pirh2. This study thus uncovers a novel function of PLAGL2 as an oncoprotein through regulating the stability of Pirh2. Given the importance of Pirh2 in regulating p53 stability, its interaction with PLAGL2 may provide valuable therapeutic targets in treating Pirh2-overexpression malignancies.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Línea Celular , Dimerización , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica
8.
Front Biosci ; 10: 975-87, 2005 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-15569636

RESUMEN

During the past decade, Akt (also known as protein kinase B, PKB) has been extensively studied. It regulates a variety of cellular processes by mediating extracellular (mitogenic growth factor, insulin and stress) and intracellular (altered tyrosine receptor kinases, Ras and Src) signals. Activation of Akt by these signals is via its pleckstrin homology (PH) domain binding to products of phosphatidylinositol 3-kinase (PI3K). This process is negatively regulated by a dual phosphatase PTEN tumor suppressor. Today, more than 30 Akt substrates have been identified. These phosphorylation events mediate the effects of Akt on cell survival, growth, differentiation, angiogenesis, migration and metabolism. Further, PI3K/PTEN/Akt pathway is frequently altered in many human malignancies and overexpression of Akt induces malignant transformation and chemoresistance. Thus, the Akt pathway is a major target for anti-cancer drug development. This review focuses on Akt signaling mechanism in oncogenesis and chemoresistance, and ongoing translational efforts to therapeutically target Akt.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Animales , Apoptosis , Ciclo Celular , Progresión de la Enfermedad , Activación Enzimática , Humanos , Isoformas de Proteínas
9.
J Cancer Res Clin Oncol ; 130(9): 521-6, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15133662

RESUMEN

PURPOSE: To investigate PRL-3 protein expression in normal colorectal epithelia and colorectal cancers with monoclonal antibody (MAb) against PRL-3. METHODS: MAb against PRL-3 was prepared with the hybridoma technique, and its specificity was confirmed with ELISA and Western blotting assays. The expression of PRL-3 protein in normal colorectal epithelia and colorectal cancers was examined by immunohistochemistry assay. Logistic regression and survival analysis were performed to determine the clinical significance of PRL-3 expression. RESULTS: MAb 3B6 against PRL-3 was obtained and showed high specificity. PRL-3 protein was expressed in two of 28 (7.1%) normal colorectal epithelia, 21 of 88 (23.9%) primary colorectal cancers, 22 of 41 (53.7%) metastatic lymph nodes and eight of 12 (66.7%) liver metastases, respectively. The PRL-3 expression rates of metastases were significantly higher than those of primary colorectal cancers and normal colorectal epithelia (P < 0.05). PRL-3 expression was significantly associated with the liver metastasis of colorectal cancer (P = 0.004) and tended to shorten survival time (P = 0.0145). CONCLUSIONS: This is the first study demonstrating that PRL-3 is a potential marker for liver metastasis of colorectal cancer and negatively influences the prognosis of colorectal cancer patients.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias Colorrectales/patología , Regulación Neoplásica de la Expresión Génica , Proteínas Inmediatas-Precoces/análisis , Neoplasias Hepáticas/química , Neoplasias Hepáticas/secundario , Proteínas Tirosina Fosfatasas/análisis , Adulto , Anciano , Análisis de Varianza , Anticuerpos Monoclonales , Western Blotting , Neoplasias Colorrectales/química , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Proteínas Inmediatas-Precoces/inmunología , Mucosa Intestinal/química , Modelos Logísticos , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias , Valor Predictivo de las Pruebas , Pronóstico , Proteínas Tirosina Fosfatasas/inmunología , Análisis de Supervivencia
10.
World J Gastroenterol ; 9(10): 2164-8, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14562370

RESUMEN

AIM: To clone and express the antigen of monoclonal antibody (MAb) PD4 for further investigation of its function. METHODS: MGC803 cDNA expression library was constructed and screened with PD4 as probes to clone the antigen. After failed in the library screening, immunoprecipitation and SDS-polyacrylamide gel electrophoresis were applied to purify the antigen for sequence analysis. The antigen coming from Mycoplasma hyorhinis (M. hyorhinis) was further confirmed with Western blot analysis by infecting M. hyorhinis -free HeLa cells and eliminating the M. hyorhinis from MGC803 cells. The full p37 gene was cloned by PCR and expressed successfully in Escherichia coli after site-directed mutations. Immunofluorescence assay was used to demonstrate if p37 protein could directly bind to gastric tumor cell AGS. RESULTS: The cDNA library constructed with MGC803 cells was screened by MAb PD4 as probes. Unfortunately, the positive clones identified with MAb PD4 were also reacted with unrelated antibodies. Then, immunoprecipitation was performed and the purified antigen was identified to be a membrane protein of Mycoplasma hyorhinis (M. hyorhinis) by sequencing of N-terminal amino acid residues. The membrane protein was intensively verified with Western blot by eliminating M. hyorhinis from MGC803 cells and by infecting M. hyorhinis-free HeLa cells. The full p37 gene was cloned and expressed successfully in Escherichia coli after site-directed mutations. Immunofluorescence demonstrated that p37 protein could directly bind to gastric tumor cell AGS. CONCLUSION: The antigen recognized by MAb PD4 is from M. hyorhinis, which suggests the actions involved in MAb PD4 is possibly mediated by p37 protein or M. hyorhinis. As p37 protein can bind directly to tumor cells, the pathogenic role of p37 involved in tumorigenesis justifies further investigation.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Escherichia coli/genética , Neoplasias Gástricas , Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos , Antígenos Bacterianos/aislamiento & purificación , Adhesión Bacteriana , Femenino , Regulación Bacteriana de la Expresión Génica/inmunología , Biblioteca de Genes , Células HeLa , Humanos , Mutagénesis Sitio-Dirigida , Infecciones por Mycoplasma/fisiopatología , Mycoplasma hyorhinis/patogenicidad , Neoplasias Ováricas , Virulencia
11.
PLoS One ; 8(4): e62729, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23658645

RESUMEN

Heterogeneous nuclear ribonucleoparticule A1/A2 (hnRNP A1/A2) and splicing factor 2/alternative splicing factor (SF2/ASF) are pivotal for precursor messenger RNA (pre-mRNA) splicing. Interferon regulatory factor-3 (IRF-3) plays critical roles in host defense against viral and microbial infection. Truncated IRF-3 proteins resulting from alternative splicing have been identified and characterized as functional antagonists to full-length IRF-3. In this study, we examined the molecular mechanism for splicing regulation of IRF-3 pre-mRNA and first reported the regulatory effect of hnRNP A1/A2 and SF2/ASF on IRF-3 splicing and activation. RNA interference-mediated depletion of hnRNP A1/A2 or SF2/ASF in human non-small cell lung cancer (NSCLC) cells increased exclusion of exons 2 and 3 of IRF-3 gene and reduced expression levels of IRF-3 protein and IRF-3 downstream effector molecules interferon-beta and CXCL10/IP-10. In addition, direct binding of hnRNP A1 and SF2/ASF to specific binding motifs in IRF-3 intron 1 was confirmed by RNA electrophoretic mobility shift assay. Subsequent minigene splicing assay showed that IRF-3 minigenes with mutated hnRNPA 1/A2 or SF2/ASF binding motifs increased exclusion of exons 2 and 3. Moreover, knockdown of hnRNP A1/A2 or SF2/ASF in NSCLC cells reinforced phytohemagglutinin-induced tumor necrosis factor-alpha release by peripheral blood mononuclear cells (PBMC) but suppressed that of interleukin-10 in NSCLC/PBMC co-cultures. Taken together, our results suggest that specific knockdown for hnRNP A1/A2 or SF2/ASF increase exclusion of exons 2 and 3 of IRF-3 pre-mRNA and influence immunomodulatory functions of human NSCLC cells.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/inmunología , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/inmunología , Factor 3 Regulador del Interferón/inmunología , Neoplasias Pulmonares/inmunología , Proteínas Nucleares/inmunología , Precursores del ARN/inmunología , Proteínas de Unión al ARN/inmunología , Empalme Alternativo , Secuencia de Bases , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Quimiocina CXCL10/genética , Quimiocina CXCL10/inmunología , Técnicas de Cocultivo , Exones , Regulación de la Expresión Génica , Ribonucleoproteína Nuclear Heterogénea A1 , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/antagonistas & inhibidores , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Humanos , Inmunomodulación , Factor 3 Regulador del Interferón/genética , Interferón beta/genética , Interferón beta/inmunología , Intrones , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Datos de Secuencia Molecular , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Precursores del ARN/genética , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/genética , Factores de Empalme Serina-Arginina , Transducción de Señal
12.
Ai Zheng ; 23(5): 602-4, 2004 May.
Artículo en Zh | MEDLINE | ID: mdl-15142464

RESUMEN

Mycoplasmas are widespread in nature as conditional pathogen, which may be the unique prokaryote that can cohabit with eukaryote and interact permanently with mammalian cells. Mycoplasma infection can be detected in many tumor tissues, continuous infection of mycoplasma can lead to transformation of mammalian cells, up-regulating expression of oncogenes, and some biologic changes of tumor cells, suggesting association of mycoplasma infection with tumorigenesis.


Asunto(s)
Infecciones por Mycoplasma , Neoplasias Ováricas/microbiología , Neoplasias Gástricas/microbiología , Animales , Transformación Celular Neoplásica , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Oncogenes , Neoplasias Ováricas/genética , Neoplasias Gástricas/genética , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA