Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Leuk Lymphoma ; 64(12): 1938-1948, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37548343

RESUMEN

Polatuzumab vedotin (Pola) was approved for first-line and relapsed/refractory (r/r) diffuse large B-cell lymphoma (DLBCL) in many countries. This means that retreatment with Pola for r/r DLBCL could be considered after first-line Pola treatment; however, there is currently no evidence on the effectiveness of Pola-retreatment. To address this, we established two Pola-resistant cells from DLBCL cells (SU-DHL-4 and STR-428) and evaluated the combination efficacy of Pola plus rituximab (Rit), the key component of DLBCL therapy. MDR1 overexpression and decreased Bim expression were suggested to be the resistant mechanisms to Pola in Pola-resistant SU-DHL-4 and Pola-resistant STR-428, respectively. In these cells, Pola significantly increased Rit-induced CDC sensitivity either with increased MAC formation or reduced Mcl-1 expression. Additionally, treatment with Pola + Rit significantly enhanced antitumor activity in Pola-resistant STR-428 xenograft mouse models. Based on these results, Pola + Rit retreatment could have preserved efficacy because of the effect of Pola on sensitizing cells to Rit.


Asunto(s)
Inmunoconjugados , Linfoma de Células B Grandes Difuso , Linfoma no Hodgkin , Humanos , Animales , Ratones , Rituximab , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/patología , Inmunoconjugados/farmacología , Inmunoconjugados/uso terapéutico , Linfoma no Hodgkin/tratamiento farmacológico , Retratamiento
2.
Br J Haematol ; 199(2): 245-255, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35764309

RESUMEN

Polatuzumab vedotin (Pola) is an antibody-drug conjugate that targets the B-cell antigen CD79b and delivers monomethyl auristatin E (MMAE). It is approved in combination with bendamustine and rituximab (Rit) for relapsed/refractory diffuse large B-cell lymphoma (r/r DLBCL). Understanding the molecular basis of Pola combination therapy with Rit, the key component for the treatment of DLBCL, is important to establish the effective treatment strategies against r/r DLBCL. Here, we examined the rationale for the combination of Pola with Rit using Pola-refractory cells. We found that treatment with Pola increased CD20 expression and sensitivity to Rit-induced complement-dependent cytotoxicity (CDC) in several Pola-refractory cells. Pola treatment increased phosphorylation of AKT and ERK and both AKT- and MEK-specific inhibitors attenuated the Pola-induced increase of CD20 and CDC sensitivity, suggesting that these phosphorylation events were required for this combination efficacy. It was revealed that anti-CD79b antibody increased the phosphorylation of AKT but inhibited the phosphorylation of ERK. In contrast, MMAE potentiated phosphorylation of ERK but slightly attenuated the phosphorylation of AKT. Pola also increased CD20 expression on Pola-refractory xenografted tumours and significantly enhanced antitumour activity in combination with Rit. In conclusion, these results could provide a novel rationale for the combination of Pola plus Rit.


Asunto(s)
Inmunoconjugados , Linfoma de Células B Grandes Difuso , Linfoma no Hodgkin , Anticuerpos Monoclonales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Clorhidrato de Bendamustina/uso terapéutico , Humanos , Inmunoconjugados/farmacología , Inmunoconjugados/uso terapéutico , Linfoma de Células B Grandes Difuso/patología , Linfoma no Hodgkin/tratamiento farmacológico , Quinasas de Proteína Quinasa Activadas por Mitógenos/uso terapéutico , Proteínas Proto-Oncogénicas c-akt , Rituximab
3.
Int J Hematol ; 115(6): 860-872, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35301681

RESUMEN

BACKGROUND: The glycoengineered, humanized anti-CD20 antibody obinutuzumab is indicated for previously untreated or relapsed/refractory CD20-positive follicular lymphoma (FL). However, the effectiveness of obinutuzumab retreatment in relapsed/refractory FL after prior obinutuzumab-containing therapy is unclear. To address this issue, we investigated the antitumor activity of obinutuzumab plus bendamustine in obinutuzumab-resistant tumors established from a human non-Hodgkin lymphoma xenograft model. MATERIALS AND METHODS: Obinutuzumab-resistant tumors (SU-DHL-4-OR-18-8) were established from an SU-DHL-4 xenograft model by repeated administration of obinutuzumab. Antitumor activity was evaluated based on tumor volume after treatment with obinutuzumab on Day 1, 8, and 15 and/or bendamustine on Day 1 and 2. Intratumoral natural killer (NK) cells/macrophages were evaluated by immunohistochemistry and flow cytometry. RESULTS: In SU-DHL-4-OR-18-8 xenografted tumors, intratumoral NK cells/macrophages after obinutuzumab treatment were significantly decreased compared with parent tumors on Day 4. The endoplasmic reticulum stress sensor phospho-IRE1 was also decreased. In SU-DHL-4-OR-18-8 tumors, bendamustine treatment increased phospho-IRE1 on Day 4 and intratumor NK cells/macrophages on Day 10. Obinutuzumab combined with bendamustine significantly increased antitumor activity compared with each single agent on Day 29, with an increase in chemoattractant CCL6 expression on Day 10. CONCLUSIONS: Coadministration of bendamustine in obinutuzumab retreatment may be effective against obinutuzumab-resistant tumors.


Asunto(s)
Linfoma Folicular , Anticuerpos Monoclonales Humanizados , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Clorhidrato de Bendamustina , Humanos , Linfoma Folicular/tratamiento farmacológico , Proteínas Serina-Treonina Quinasas , Rituximab
4.
Mol Biol Rep ; 49(6): 4421-4433, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35218445

RESUMEN

BACKGROUND: Obinutuzumab, a Type II anti-CD20 antibody, is used to treat follicular lymphoma. A major mode of action of obinutuzumab is antibody-dependent cellular cytotoxicity (ADCC). Knowledge of the mechanisms of resistance to obinutuzumab is important for the development of next-line strategies to follow obinutuzumab-containing therapy, including obinutuzumab retreatment. Unfortunately, the mechanisms by which tumor cells acquire resistance to ADCC are still poorly understood. To address this, we examined the mechanisms of resistance to obinutuzumab-induced ADCC and the combination efficacy of obinutuzumab and clinically available agents in the established resistant cells. METHODS AND RESULTS: We established cells resistant to obinutuzumab-induced ADCC using the non-Hodgkin lymphoma cell line RL and examined their mechanisms of resistance and the combination efficacy of obinutuzumab and clinically available agents. Comprehensive analysis by RNA sequencing of resistance mechanisms revealed that abnormal Fas signaling decreased sensitivity to ADCC in resistant clones. Combination treatment with prednisolone, a component of CHOP and CVP, was found to enhance ADCC sensitivity of RL cells and resistant clones and to significantly suppress tumor growth in xenograft models. Treatment with prednisolone upregulated expression of CD20 and an apoptosis-inducing protein BIM, which might augment perforin/granzyme B-mediated cell death. Furthermore, pretreatment of the effector cells with bendamustine enhanced ADCC activity, and treatment with obinutuzumab plus bendamustine showed significant antitumor efficacy in xenograft models. It was speculated that bendamustine upregulates ADCC activity by potentiating granules-mediated cell killing. CONCLUSIONS: Our study revealed a novel mechanism underlying obinutuzumab-induced ADCC resistance and indicated that ADCC resistance could be overcome by combining obinutuzumab with prednisolone or bendamustine. This study provides a scientific rationale for obinutuzumab-retreatment in combination with clinically available chemotherapeutic agents for obinutuzumab resistant follicular lymphoma.


Asunto(s)
Linfoma Folicular , Anticuerpos Monoclonales Humanizados , Citotoxicidad Celular Dependiente de Anticuerpos , Clorhidrato de Bendamustina/uso terapéutico , Humanos , Linfoma Folicular/tratamiento farmacológico , Linfoma Folicular/patología , Prednisolona , Rituximab/farmacología , Rituximab/uso terapéutico
5.
Mol Cancer Ther ; 20(6): 1133-1141, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33850006

RESUMEN

Follicular lymphoma commonly recurs and is difficult to cure. Obinutuzumab is a humanized glycoengineered type II anti-CD20 antibody with a mode of action that includes induction of antibody-dependent cellular cytotoxicity, antibody-dependent cellular phagocytosis, and direct cell death. There is no evidence on the effectiveness of retreatment with obinutuzumab in patients with prior obinutuzumab treatment. Using obinutuzumab-induced direct-cell-death-resistant cells, we investigated the efficacy of obinutuzumab retreatment in combination with chemotherapeutic agents used in follicular lymphoma treatment. Human non-Hodgkin lymphoma SU-DHL-4 cells were sustainably exposed to obinutuzumab in vitro, and 17 resistant clones expressing CD20 and showing 100-fold higher IC50 of obinutuzumab than parental cells were established. The growth inhibition effect of obinutuzumab in combination with bendamustine, 4-hydroperoxy-cyclophosphamide, doxorubicin, vincristine, or prednisolone was estimated using an interaction index based on the Bliss independence model. For each clone, there were various combinations of obinutuzumab and chemotherapeutic agents that showed supra-additive effects. Obinutuzumab combined with doxorubicin enhanced caspase-dependent apoptosis and growth inhibition effect. Obinutuzumab combined with prednisolone enhanced DNA fragmentation and G0-G1 arrest. These combinations also had an antitumor effect in mouse xenograft models. Our results indicate that retreatment with obinutuzumab, when it is combined with chemotherapeutic agents, is effective in the CD20-positive obinutuzumab-induced direct-cell-death-resistant cells.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Muerte Celular/efectos de los fármacos , Linfoma no Hodgkin/tratamiento farmacológico , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos Inmunológicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones
6.
J Nutr Sci Vitaminol (Tokyo) ; 64(4): 271-276, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30175790

RESUMEN

Abdominal aortic aneurysm (AAA) is a vascular disease characterized by chronic inflammation in the infrarenal aorta. Epidemiologic data have clearly linked tobacco smoking to aneurysm formation and a faster rate of expansion. It suggested that nicotine, one of the main ingredients of tobacco, has been suggested to be associated with AAA development and rupture. In the condition where no established drugs are available; therefore, an effective approach to prevent the vascular damage from nicotine consumption may be the use of dietary functional food factors. However, little is known about the relationship between dietary components and AAA. In this study, we estimated the effect of dietary deoxyribonucleic acid (DNA) on the vascular wall. After habituation for 5 d, the mice were divided into four groups: control diet and distilled water group (C), DNA-Na diet and distilled water group (DNA), control diet and 0.5 mg/mL nicotine solution group (C-Nic), DNA-Na diet, and 0.5 mg/mL nicotine solution group (DNA-Nic). The dietary DNA attenuated the degradation of elastin fibers induced by nicotine administration. The areas stained positive for MMP-2 in the DNA-Nic group were significantly suppressed compared to C-Nic mice. These data suggest that the dietary DNA may prevent the weakening of the aortic wall via inhibition of the MMP-2-dependent pathway. In conclusion, we have revealed the protective effect of dietary DNA on the vascular pathology of nicotine-administrated mice. A nucleic acid-rich diet might be useful for people who consume nicotine via smoking, chewing tobacco, or nicotine patches.


Asunto(s)
Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/prevención & control , ADN/uso terapéutico , Suplementos Dietéticos , Modelos Animales de Enfermedad , Elastina/metabolismo , Endotelio Vascular/metabolismo , Adventicia/efectos de los fármacos , Adventicia/inmunología , Adventicia/metabolismo , Adventicia/patología , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Antioxidantes/uso terapéutico , Aorta Abdominal/efectos de los fármacos , Aorta Abdominal/inmunología , Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/inducido químicamente , Aneurisma de la Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/patología , Fármacos Cardiovasculares/uso terapéutico , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/inmunología , Endotelio Vascular/patología , Inmunohistoquímica , Masculino , Metaloproteinasa 2 de la Matriz/química , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones Endogámicos C57BL , Nicotina/toxicidad , Estrés Oxidativo/efectos de los fármacos , Proteolisis/efectos de los fármacos
7.
Cancer Sci ; 109(7): 2211-2220, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29722104

RESUMEN

Long noncoding RNAs are involved in a variety of cellular functions. In particular, an increasing number of studies have revealed the functions of long noncoding RNA in various cancers; however, their precise roles and mechanisms of action remain to be elucidated. NORAD, a cytoplasmic long noncoding RNA, is upregulated by irradiation and functions as a potential oncogenic factor by binding and inhibiting Pumilio proteins (PUM1/PUM2). Here, we show that NORAD upregulates transforming growth factor-ß (TGF-ß) signaling and regulates TGF-ß-induced epithelial-to-mesenchymal transition (EMT)-like phenotype, which is a critical step in the progression of lung adenocarcinoma, A549 cells. However, PUM1 does not appear to be involved in this process. We thus focused on importin ß1 as a binding partner of NORAD and found that knockdown of NORAD partially inhibits the physical interaction of importin ß1 with Smad3, inhibiting the nuclear accumulation of Smad complexes in response to TGF-ß. Our findings may provide a new mechanism underlying the function of NORAD in cancer cells.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Transición Epitelial-Mesenquimal/fisiología , Neoplasias Pulmonares/patología , ARN Largo no Codificante/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Células A549 , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Fenotipo , Transducción de Señal
8.
Cell Discov ; 4: 1, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29423269

RESUMEN

The mammalian target of rapamycin (mTOR) pathway is commonly activated in human cancers. The activity of mTOR complex 1 (mTORC1) signaling is supported by the intracellular positioning of cellular compartments and vesicle trafficking, regulated by Rab GTPases. Here we showed that tuftelin 1 (TUFT1) was involved in the activation of mTORC1 through modulating the Rab GTPase-regulated process. TUFT1 promoted tumor growth and metastasis. Consistently, the expression of TUFT1 correlated with poor prognosis in lung, breast and gastric cancers. Mechanistically, TUFT1 physically interacted with RABGAP1, thereby modulating intracellular lysosomal positioning and vesicular trafficking, and promoted mTORC1 signaling. In addition, expression of TUFT1 predicted sensitivity to perifosine, an alkylphospholipid that alters the composition of lipid rafts. Perifosine treatment altered the positioning and trafficking of cellular compartments to inhibit mTORC1. Our observations indicate that TUFT1 is a key regulator of the mTORC1 pathway and suggest that it is a promising therapeutic target or a biomarker for tumor progression.

9.
Nucleic Acids Res ; 46(3): 1180-1195, 2018 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-29186616

RESUMEN

It is well established that transforming growth factor-ß (TGFß) switches its function from being a tumor suppressor to a tumor promoter during the course of tumorigenesis, which involves both cell-intrinsic and environment-mediated mechanisms. We are interested in breast cancer cells, in which SMAD mutations are rare and interactions between SMAD and other transcription factors define pro-oncogenic events. Here, we have performed chromatin immunoprecipitation (ChIP)-sequencing analyses which indicate that the genome-wide landscape of SMAD2/3 binding is altered after prolonged TGFß stimulation. De novo motif analyses of the SMAD2/3 binding regions predict enrichment of binding motifs for activator protein (AP)1 in addition to SMAD motifs. TGFß-induced expression of the AP1 component JUNB was required for expression of many late invasion-mediating genes, creating a feed-forward regulatory network. Moreover, we found that several components in the WNT pathway were enriched among the late TGFß-target genes, including the invasion-inducing WNT7 proteins. Consistently, overexpression of WNT7A or WNT7B enhanced and potentiated TGFß-induced breast cancer cell invasion, while inhibition of the WNT pathway reduced this process. Our study thereby helps to explain how accumulation of pro-oncogenic stimuli switches and stabilizes TGFß-induced cellular phenotypes of epithelial cells.


Asunto(s)
Neoplasias de la Mama/genética , Retroalimentación Fisiológica , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Factores de Transcripción/genética , Factor de Crecimiento Transformador beta1/farmacología , Animales , Secuencia de Bases , Sitios de Unión , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular , Línea Celular Tumoral , Embrión no Mamífero , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Invasividad Neoplásica , Unión Proteica , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína smad3/genética , Proteína smad3/metabolismo , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , Pez Cebra
10.
Sci Rep ; 7(1): 1166, 2017 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-28446749

RESUMEN

Epithelial-mesenchymal transition (EMT) is induced by transforming growth factor (TGF)-ß and facilitates tumor progression. We here performed global mapping of accessible chromatin in the mouse mammary gland epithelial EpH4 cell line and its Ras-transformed derivative (EpRas) using formaldehyde-assisted isolation of regulatory element (FAIRE)-sequencing. TGF-ß and Ras altered chromatin accessibility either cooperatively or independently, and AP1, ETS, and RUNX binding motifs were enriched in the accessible chromatin regions of EpH4 and EpRas cells. Etv4, an ETS family oncogenic transcription factor, was strongly expressed and bound to more than one-third of the accessible chromatin regions in EpRas cells treated with TGF-ß. While knockdown of Etv4 and another ETS family member Etv5 showed limited effects on the decrease in the E-cadherin abundance and stress fiber formation by TGF-ß, gene ontology analysis showed that genes encoding extracellular proteins were most strongly down-regulated by Etv4 and Etv5 siRNAs. Accordingly, TGF-ß-induced expression of Mmp13 and cell invasiveness were suppressed by Etv4 and Etv5 siRNAs, which were accompanied by the reduced chromatin accessibility at an enhancer region of Mmp13 gene. These findings suggest a mechanism of transcriptional regulation during Ras- and TGF-ß-induced EMT that involves alterations of accessible chromatin, which are partly regulated by Etv4 and Etv5.


Asunto(s)
Transformación Celular Neoplásica , Cromatina/metabolismo , Células Epiteliales/fisiología , Transición Epitelial-Mesenquimal , Glándulas Mamarias Animales/citología , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Sitios de Unión , Línea Celular , ADN/metabolismo , Regulación de la Expresión Génica , Ratones , Unión Proteica
11.
Sci Signal ; 9(442): ra84, 2016 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-27555661

RESUMEN

The p53 family of transcription factors includes p63, which is a master regulator of gene expression in epithelial cells. Determining whether p63 is tumor-suppressive or tumorigenic is complicated by isoform-specific and cellular context-dependent protein associations, as well as antagonism from mutant p53. ΔNp63 is an amino-terminal-truncated isoform, that is, the predominant isoform expressed in cancer cells of epithelial origin. In HaCaT keratinocytes, which have mutant p53 and ΔNp63, we found that mutant p53 antagonized ΔNp63 transcriptional activity but that activation of Ras or transforming growth factor-ß (TGF-ß) signaling pathways reduced the abundance of mutant p53 and strengthened target gene binding and activity of ΔNp63. Among the products of ΔNp63-induced genes was dual-specificity phosphatase 6 (DUSP6), which promoted the degradation of mutant p53, likely by dephosphorylating p53. Knocking down all forms of p63 or DUSP6 and DUSP7 (DUSP6/7) inhibited the basal or TGF-ß-induced or epidermal growth factor (which activates Ras)-induced migration and invasion in cultures of p53-mutant breast cancer and squamous skin cancer cells. Alternatively, overexpressing ΔNp63 in the breast cancer cells increased their capacity to colonize various tissues upon intracardiac injection in mice, and this was inhibited by knocking down DUSP6/7 in these ΔNp63-overexpressing cells. High abundance of ΔNp63 in various tumors correlated with poor prognosis in patients, and this correlation was stronger in patients whose tumors also had a mutation in the gene encoding p53. Thus, oncogenic Ras and TGF-ß signaling stimulate cancer progression through activation of the ΔNp63 transcriptional program.


Asunto(s)
Neoplasias de la Mama/metabolismo , Movimiento Celular , Proteína Oncogénica p21(ras)/metabolismo , Transducción de Señal , Transcripción Genética , Factor de Crecimiento Transformador beta/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Transformada , Fosfatasa 6 de Especificidad Dual/genética , Fosfatasa 6 de Especificidad Dual/metabolismo , Femenino , Células HEK293 , Humanos , Proteína Oncogénica p21(ras)/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
12.
Stem Cell Reports ; 6(1): 64-73, 2016 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-26771354

RESUMEN

Bone morphogenetic protein (BMP) signaling exerts paradoxical roles in pluripotent stem cells (PSCs); it sustains self-renewal of mouse embryonic stem cells (ESCs), while it induces differentiation in other PSCs, including human ESCs. Here, we revisit the roles of BMP-4 using mouse ESCs (mESCs) in naive and primed states. SMAD1 and SMAD5, which transduce BMP signals, recognize enhancer regions together with KLF4 and KLF5 in naive mESCs. KLF4 physically interacts with SMAD1 and suppresses its activity. Consistently, a subpopulation of cells with active BMP-SMAD can be ablated without disturbing the naive state of the culture. Moreover, Smad1/5 double-knockout mESCs stay in the naive state, indicating that the BMP-SMAD pathway is dispensable for it. In contrast, the MEK5-ERK5 pathway mediates BMP-4-induced self-renewal of mESCs by inducing Klf2, a critical factor for the ground state pluripotency. Our study illustrates that BMP exerts its self-renewing effect through distinct functions of different Krüppel-like factors.


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , Autorrenovación de las Células/fisiología , Factores de Transcripción de Tipo Kruppel/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Animales , Western Blotting , Proteína Morfogenética Ósea 4/genética , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Autorrenovación de las Células/genética , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Células Hep G2 , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Sistema de Señalización de MAP Quinasas/genética , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Noqueados , Células Madre Embrionarias de Ratones/citología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Unión Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína Smad1/genética , Proteína Smad1/metabolismo , Proteína Smad5/genética , Proteína Smad5/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...