Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Mol Cancer ; 23(1): 45, 2024 02 29.
Artículo en Inglés | MEDLINE | ID: mdl-38424542

RESUMEN

BACKGROUND: In the myeloid compartment of the tumor microenvironment, CD244 signaling has been implicated in immunosuppressive phenotype of monocytes. However, the precise molecular mechanism and contribution of CD244 to tumor immunity in monocytes/macrophages remains elusive due to the co-existing lymphoid cells expressing CD244. METHODS: To directly assess the role of CD244 in tumor-associated macrophages, monocyte-lineage-specific CD244-deficient mice were generated using cre-lox recombination and challenged with B16F10 melanoma. The phenotype and function of tumor-infiltrating macrophages along with antigen-specific CD8 T cells were analyzed by flow cytometry and single cell RNA sequencing data analysis, and the molecular mechanism underlying anti-tumorigenic macrophage differentiation, antigen presentation, phagocytosis was investigated ex vivo. Finally, the clinical feasibility of CD244-negative monocytes as a therapeutic modality in melanoma was confirmed by adoptive transfer experiments. RESULTS: CD244fl/flLysMcre mice demonstrated a significant reduction in tumor volume (61% relative to that of the CD244fl/fl control group) 14 days after tumor implantation. Within tumor mass, CD244fl/flLysMcre mice also showed higher percentages of Ly6Clow macrophages, along with elevated gp100+IFN-γ+ CD8 T cells. Flow cytometry and RNA sequencing data demonstrated that ER stress resulted in increased CD244 expression on monocytes. This, in turn, impeded the generation of anti-tumorigenic Ly6Clow macrophages, phagocytosis and MHC-I antigen presentation by suppressing autophagy pathways. Combining anti-PD-L1 antibody with CD244-/- bone marrow-derived macrophages markedly improved tumor rejection compared to the anti-PD-L1 antibody alone or in combination with wild-type macrophages. Consistent with the murine data, transcriptome analysis of human melanoma tissue single-cell RNA-sequencing dataset revealed close association between CD244 and the inhibition of macrophage maturation and function. Furthermore, the presence of CD244-negative monocytes/macrophages significantly increased patient survival in primary and metastatic tumors. CONCLUSION: Our study highlights the novel role of CD244 on monocytes/macrophages in restraining anti-tumorigenic macrophage generation and tumor antigen-specific T cell response in melanoma. Importantly, our findings suggest that CD244-deficient macrophages could potentially be used as a therapeutic agent in combination with immune checkpoint inhibitors. Furthermore, CD244 expression in monocyte-lineage cells serve as a prognostic marker in cancer patients.


Asunto(s)
Melanoma , Monocitos , Humanos , Animales , Ratones , Monocitos/metabolismo , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/metabolismo , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Macrófagos/metabolismo , Linfocitos T CD8-positivos , Carcinogénesis/metabolismo , Microambiente Tumoral , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo
2.
Infect Immun ; 89(4)2021 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-33468581

RESUMEN

Thymosin beta-4 (Tß4) is an actin-sequestering peptide that plays important roles in regeneration and remodeling of injured tissues. However, its function in a naturally occurring pathogenic bacterial infection model has remained elusive. We adopted Tß4-overexpressing transgenic (Tg) mice to investigate the role of Tß4 in acute pulmonary infection and systemic sepsis caused by Legionella pneumophila Upon infection, Tß4-Tg mice demonstrated significantly lower bacterial loads in the lung, less hyaline membranes and necrotic abscess, with lower interstitial infiltration of neutrophils, CD4+, and CD8+ T cells. Bronchoalveolar lavage fluid of Tß4-Tg mice possessed higher bactericidal activity against exogenously added L. pneumophila, suggesting that constitutive expression of Tß4 could efficiently control L. pneumophila Furthermore, qPCR analysis of lung homogenates demonstrated significant reduction of interleukin 1 beta (IL-1ß) and tumor necrosis factor alpha (TNF-α), which primarily originate from lung macrophages, in Tß4-Tg mice after pulmonary infection. Upon L. pneumophila challenge of bone marrow-derived macrophages (BMDM) in vitro, secretion of IL-1ß and TNF-α proteins was also reduced in Tß4-Tg macrophages, without affecting their survival. The anti-inflammatory effects of BMDM in Tß4-Tg mice on each cytokine were affected when triggering with tlr2, tlr4, tlr5, or tlr9 ligands, suggesting that anti-inflammatory effects of Tß4 are likely mediated by the reduced activation of Toll-like receptors (TLR). Finally, Tß4-Tg mice in a systemic sepsis model were protected from L. pneumophila-induced lethality compared to wild-type controls. Therefore, Tß4 confers effective resistance against L. pneumophila via two pathways, a bactericidal and an anti-inflammatory pathway, which can be harnessed to treat acute pneumonia and septic conditions caused by L. pneumophila in humans.


Asunto(s)
Resistencia a la Enfermedad/genética , Expresión Génica Ectópica , Legionella pneumophila/fisiología , Enfermedad de los Legionarios/genética , Enfermedad de los Legionarios/microbiología , Neumonía Bacteriana/genética , Neumonía Bacteriana/microbiología , Timosina/genética , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Interacciones Huésped-Patógeno/genética , Humanos , Inmunohistoquímica , Inmunofenotipificación , Enfermedad de los Legionarios/patología , Ligandos , Masculino , Ratones , Ratones Transgénicos , Neumonía Bacteriana/patología , Sepsis/genética , Sepsis/microbiología , Sepsis/patología , Receptores Toll-Like/metabolismo
3.
Int J Mol Sci ; 21(7)2020 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-32260561

RESUMEN

The recent development of high-throughput genomics has revolutionized personalized medicine by identifying key pathways and molecular targets controlling tumor progression and survival. Mitogen-activated protein kinase (MAPK) pathways are examples of such targets, and inhibitors against these pathways have shown promising clinical responses in patients with melanoma, non-small-cell lung cancer, colorectal cancer, pancreatic cancer, and thyroid cancer. Although MAPK pathway-targeted therapies have resulted in significant clinical responses in a large proportion of cancer patients, the rate of tumor recurrence is high due to the development of resistance. Conversely, immunotherapies have shown limited clinical responses, but have led to durable tumor regression in patients, and complete responses. Recent evidence indicates that MAPK-targeted therapies may synergize with immune cells, thus providing rationale for the development of combination therapies. Here, we review the current status of ongoing clinical trials investigating MAPK pathway inhibitors, such as BRAF and MAPK/ERK kinase (MEK) inhibitors, in combination with checkpoint inhibitors targeting programmed death protein 1 (PD-1), programmed death-ligand 1 (PD-L1), and cytotoxic T cell associated antigen-4 (CTLA-4). A better understanding of an individual drug's mechanism of action, patterns of acquired resistance, and the influence on immune cells will be critical for the development of novel combination therapies.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia/métodos , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Animales , Ensayos Clínicos como Asunto , Terapia Combinada , Humanos , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Neoplasias/terapia
4.
PLoS Genet ; 12(2): e1005884, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26925584

RESUMEN

The inactivation of p53 creates a major challenge for inducing apoptosis in cancer cells. An attractive strategy is to identify and subsequently target the survival signals in p53 defective cancer cells. Here we uncover a RUNX2-mediated survival signal in p53 defective cancer cells. The inhibition of this signal induces apoptosis in cancer cells but not non-transformed cells. Using the CRISPR technology, we demonstrate that p53 loss enhances the apoptosis caused by RUNX2 knockdown. Mechanistically, RUNX2 provides the survival signal partially through inducing MYC transcription. Cancer cells have high levels of activating histone marks on the MYC locus and concomitant high MYC expression. RUNX2 knockdown decreases the levels of these histone modifications and the recruitment of the Menin/MLL1 (mixed lineage leukemia 1) complex to the MYC locus. Two inhibitors of the Menin/MLL1 complex induce apoptosis in p53 defective cancer cells. Together, we identify a RUNX2-mediated epigenetic mechanism of the survival of p53 defective cancer cells and provide a proof-of-principle that the inhibition of this epigenetic axis is a promising strategy to kill p53 defective cancer cells.


Asunto(s)
Neoplasias Óseas/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Epigénesis Genética , Osteosarcoma/genética , Proteína p53 Supresora de Tumor/genética , Animales , Apoptosis/genética , Sitios de Unión , Neoplasias Óseas/patología , Línea Celular Tumoral , Supervivencia Celular/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad beta del Factor de Unión al Sitio Principal/genética , Subunidad beta del Factor de Unión al Sitio Principal/metabolismo , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Genes myc , Humanos , Ratones Noqueados , Osteosarcoma/patología , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Stem Cells ; 33(4): 1304-19, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25524638

RESUMEN

The tumor suppressor, p53, plays a critical role in suppressing osteosarcoma. Bone marrow stromal cells (BMSCs, also known as bone marrow-derived mesenchymal stem cells) have been suggested to give rise to osteosarcomas. However, the role of p53 in BMSCs has not been extensively explored. Here, we report that p53 regulates the lineage choice of mouse BMSCs (mBMSCs). Compared to mBMSCs with wild-type p53, mBMSCs deficient in p53 have enhanced osteogenic differentiation, but with similar adipogenic and chondrogenic differentiation. The role of p53 in inhibiting osteogenic lineage differentiation is mainly through the action of Runx2, a master transcription factor required for the osteogenic differentiation of mBMSCs. We find that p53 indirectly represses the expression of Runx2 by activating the microRNA-34 family, which suppresses the translation of Runx2. Since osteosarcoma may derive from BMSCs, we examined whether p53 has a role in the osteogenic differentiation of osteosarcoma cells and found that osteosarcoma cells with p53 deletion have higher levels of Runx2 and faster osteogenic differentiation than those with wild-type p53. A systems biology approach reveals that p53-deficient mBMSCs are more closely related to human osteosarcoma while mBMSCs with wild-type p53 are similar to normal human BMSCs. In summary, our results indicate that p53 activity can influence cell fate specification of mBMSCs, and provide molecular and cellular insights into the observation that p53 loss is associated with increased osteosarcoma incidence.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Mesenquimatosas/metabolismo , Osteogénesis/fisiología , Proteína p53 Supresora de Tumor/deficiencia , Animales , Línea Celular Tumoral , Células Cultivadas , Humanos , Ratones , Ratones Noqueados
6.
Cells ; 12(12)2023 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-37371075

RESUMEN

Adoptive cell therapy using chimeric antigen receptor (CAR) technology is one of the most advanced engineering platforms for cancer immunotherapy. CAR-T cells have shown remarkable efficacy in the treatment of hematological malignancies. However, their limitations in solid tumors include an immunosuppressive tumor microenvironment (TME), insufficient tumor infiltration, toxicity, and the absence of tumor-specific antigens. Although recent advances in CAR-T cell design-such as the incorporation of co-stimulatory domains and the development of armored CAR-T cells-have shown promising results in treating solid tumors, there are still challenges that need to be addressed. To overcome these limitations, other immune cells, such as natural killer (NK) cells and macrophages (M), have been developed as attractive options for efficient cancer immunotherapy of solid tumors. CAR-NK cells exhibit substantial clinical improvements with "off-the-shelf" availability and low toxicity. CAR-M cells have promising therapeutic potential because macrophages can infiltrate the TME of solid tumors. Here, we review the recent advances and future perspectives associated with engineered immune cell-based cancer immunotherapies for solid tumors. We also summarize ongoing clinical trials investigating the safety and efficacy of engineered immune cells, such as CAR-T, CAR-NK, and CAR-M, for targeting solid tumors.


Asunto(s)
Neoplasias , Receptores Quiméricos de Antígenos , Humanos , Receptores Quiméricos de Antígenos/metabolismo , Inmunoterapia Adoptiva/métodos , Neoplasias/patología , Inmunoterapia/métodos , Linfocitos T , Antígenos de Neoplasias/metabolismo , Microambiente Tumoral
7.
Biomed Pharmacother ; 154: 113553, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35994815

RESUMEN

To overcome the hurdles of immunotherapy, we investigated whether calcipotriol, a synthetic vitamin D analog, could overcome the immune evasion of glioblastoma multiforme (GBM) by modulating immune responses and the immunosuppressive tumor microenvironment. Administration of calcipotriol considerably reduced tumor growth. Both in vivo and in vitro studies revealed that CD8+T and natural killer (NK) cell gene signatures were enriched and activated, producing high levels of IFN-γ and granzyme B. In contrast, regulatory T cells (Treg) were significantly reduced in the calcipotriol-treated group. The expression of CD127, the receptor for thymic stromal lymphopoietin (TSLP), is elevated in CD4+T cells and potentially supports T-cell priming. Depleting CD4+T cells, but not NK or CD8+T cells, completely abrogated the antitumor efficacy of calcipotriol. These data highlight that the calcipotriol/TSLP/CD4+T axis can activate CD8+T and NK cells with a concomitant reduction in the number of Tregs in GBM. Therefore, calcipotriol can be a novel therapeutic modality to overcome the immune resistance of GBM by converting immunologically "cold" tumors into "hot" tumors. DATA AVAILABILITY: Data are available upon reasonable request. The RNA-seq dataset comparing the transcriptomes of control and calcipotriol-treated GL261 tumors is available from the corresponding author upon request.


Asunto(s)
Glioblastoma , Vitamina D , Linfocitos T CD8-positivos , Calcitriol/análogos & derivados , Glioblastoma/metabolismo , Humanos , Células Asesinas Naturales , Activación de Linfocitos , Microambiente Tumoral , Vitamina D/metabolismo
8.
Cancers (Basel) ; 13(8)2021 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-33920906

RESUMEN

NK cells are the predominant innate lymphocyte subsets specialized to kill malignant tumor cells. In patients with advanced cancer, hypoxic stress shapes NK cells toward tumor-resistant and immunosuppressive phenotypes, hence a strategy to restore NK function is critical for successful tumor immunotherapy. Here, we present evidence that pre-activation and subsequent HIF-1α-dependent metabolic shift of NK cells from oxidative phosphorylation into glycolysis are keys to overcome hypoxia-mediated impairment in NK cell survival, proliferation, and tumor cytotoxicity. Specifically, exposing NK cells to 7-9 days of normoxic culture followed by a pO2 of 1.5% hypoxia led to a highly potent effector phenotype via HIF-1α stabilization and upregulation of its target genes, BNIP3, PDK1, VEGF, PKM2, and LDHA. RNA sequencing and network analyses revealed that concomitant reduction of p21/p53 apoptotic pathways along with upregulation of cell cycle-promoting genes, CCNE1, CDC6, CDC20, and downregulation of cell cycle-arrest genes, CDKN1A, GADD45A, and MDM2 were accountable for superior expansion of NK cells via ERK/STAT3 activation. Furthermore, HIF-1α-dependent upregulation of the NKp44 receptor in hypoxia-exposed NK cells resulted in increased killing against K562, CEM, and A375 tumor targets both in-vitro and in-vivo tumor clearance assays. Therefore, hypoxic exposure on pre-activated proliferating NK cells triggered HIF-1α-dependent pathways to initiate coordinated regulation of cell cycle, apoptosis, and cytotoxicity at the global gene transcription level. Our results uncover a previously unidentified role of HIF-1α-mediated metabolic reprogramming that can reverse impaired NK effector phenotypes to generate requisite numbers of functionally robust NK cells for adoptive cellular therapy for clinical evaluation.

9.
Immune Netw ; 20(2): e14, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32395366

RESUMEN

With the development of technologies that can transform immune cells into therapeutic modalities, immunotherapy has remarkably changed the current paradigm of cancer treatment in recent years. NK cells are components of the innate immune system that act as key regulators and exhibit a potent tumor cytolytic function. Unlike T cells, NK cells exhibit tumor cytotoxicity by recognizing non-self, without deliberate immunization or activation. Currently, researchers have developed various approaches to improve the number and anti-tumor function of NK cells. These approaches include the use of cytokines and Abs to stimulate the efficacy of NK cell function, adoptive transfer of autologous or allogeneic ex vivo expanded NK cells, establishment of homogeneous NK cell lines using the NK cells of patients with cancer or healthy donors, derivation of NK cells from induced pluripotent stem cells (iPSCs), and modification of NK cells with cutting-edge genetic engineering technologies to generate chimeric Ag receptor (CAR)-NK cells. Such NK cell-based immunotherapies are currently reported as being promising anti-tumor strategies that have shown enhanced functional specificity in several clinical trials investigating malignant tumors. Here, we summarize the recent advances in NK cell-based cancer immunotherapies that have focused on providing improved function through the use of the latest genetic engineering technologies. We also discuss the different types of NK cells developed for cancer immunotherapy and present the clinical trials being conducted to test their safety and efficacy.

10.
Mol Cell Biol ; 26(23): 8826-39, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16982676

RESUMEN

Mechanisms coordinating neural progenitor cell cycle exit and differentiation are incompletely understood. The cyclin-dependent kinase inhibitor p27(Kip1) is transcriptionally induced, switching specific neural progenitors from proliferation to differentiation. However, neuronal differentiation-specific transcription factors mediating p27(Kip1) transcription have not been identified. We demonstrate the homeodomain transcription factor Phox2a, required for central nervous system (CNS)- and neural crest (NC)-derived noradrenergic neuron differentiation, coordinates cell cycle exit and differentiation by inducing p27(Kip1) transcription. Phox2a transcription and activation in the CNS-derived CAD cell line and primary NC cells is mediated by combined cyclic AMP (cAMP) and bone morphogenetic protein 2 (BMP2) signaling. In the CAD cellular model, cAMP and BMP2 signaling initially induces proliferation of the undifferentiated precursors, followed by p27(Kip1) transcription, G(1) arrest, and neuronal differentiation. Small interfering RNA silencing of either Phox2a or p27(Kip1) suppresses p27(Kip1) transcription and neuronal differentiation, suggesting a causal link between p27(Kip1) expression and differentiation. Conversely, ectopic Phox2a expression via the Tet-off expression system promotes accelerated CAD cell neuronal differentiation and p27(Kip1) transcription only in the presence of cAMP signaling. Importantly, endogenous or ectopically expressed Phox2a activated by cAMP signaling binds homeodomain cis-acting elements of the p27(Kip1) promoter in vivo and mediates p27(Kip1)-luciferase expression in CAD and NC cells. We conclude that developmental cues of cAMP signaling causally link Phox2a activation with p27(Kip1) transcription, thereby coordinating neural progenitor cell cycle exit and differentiation.


Asunto(s)
Ciclo Celular/fisiología , Diferenciación Celular/fisiología , AMP Cíclico/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Proteínas de Homeodominio/genética , Cresta Neural/embriología , Células Madre/citología , Transcripción Genética , Animales , Línea Celular , Células Cultivadas , Inmunoprecipitación de Cromatina , Coturnix/embriología , AMP Cíclico/fisiología , Inmunohistoquímica , Modelos Biológicos , Cresta Neural/citología , Cresta Neural/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transfección
11.
Front Immunol ; 10: 496, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31024520

RESUMEN

Tumor-infiltrating lymphocytes (TILs), found in patients with advanced pancreatic ductal adenocarcinoma (PDAC), are shown to correlate with overall survival (OS) rate. Although majority of TILs consist of CD8+/CD4+ T cells, the presence of NK cells and their role in the pathogenesis of PDAC remains elusive. We performed comprehensive analyses of TIL, PBMC, and autologous tumor cells from 80 enrolled resectable PDAC patients to comprehend the NK cell defects within PDAC. Extremely low frequencies of NK cells (<0.5%) were found within PDAC tumors, which was attributable not to the low expression of tumor chemokines, but to the lack of chemokine receptor, CXCR2. Forced expression of CXCR2 in patients' NK cells rendered them capable of trafficking into PDAC. Furthermore, NK cells exhibited impaired cell-mediated killing of autologous PDAC cells, primarily due to insufficient ligation of NKG2D and DNAM-1, and failed to proliferate within the hypoxic tumor microenvironment. Importantly, these defects could be overcome by ex-vivo stimulation of NK cells from such patients. Importantly, when the proliferative capacity of NK cells in vitro was used to stratify patients on the basis of cell expansion, patients whose NK cells proliferated <250-fold experienced significantly lower DFS and OS than those with ≥250-fold. Ex-vivo activation of NK cells restored tumor trafficking and reactivity, hence provided a therapeutic modality while their fold expansion could be a potentially significant prognostic indicator of OS and DFS in such patients.


Asunto(s)
Células Asesinas Naturales/inmunología , Neoplasias Pancreáticas/inmunología , Escape del Tumor/inmunología , Adenocarcinoma/inmunología , Anciano , Carcinoma Ductal Pancreático/inmunología , Línea Celular Tumoral , Proliferación Celular/fisiología , Femenino , Humanos , Células K562 , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Neoplasias Pancreáticas/patología , Pronóstico , Microambiente Tumoral/inmunología
12.
Nat Commun ; 10(1): 3258, 2019 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-31332204

RESUMEN

CD160 and BTLA both bind to herpes virus entry mediator. Although a negative regulatory function of BTLA in natural killer T (NKT) cell activation has been reported, whether CD160 is also involved is unclear. By analyzing CD160-/- mice and mixed bone marrow chimeras, we show that CD160 is not essential for NKT cell development. However, CD160-/- mice exhibit severe liver injury after in vivo challenge with α-galactosylceramide (α-GalCer). Moreover, CD160-/- mice are more susceptible to Concanavalin A challenge, and display elevated serum AST and ALT levels, hyperactivation of NKT cells, and enhanced IFN-γ, TNF, and IL-4 production. Lastly, inhibition of BTLA by anti-BTLA mAb aggravates α-GalCer-induced hepatic injury in CD160-/- mice, suggesting that both CD160 and BTLA serve as non-overlapping negative regulators of NKT cells. Our data thus implicate CD160 as a co-inhibitory receptor that delivers antigen-dependent signals in NKT cells to dampen cytokine production during early innate immune activation.


Asunto(s)
Antígenos CD/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Hígado/metabolismo , Células T Asesinas Naturales/metabolismo , Receptores Inmunológicos/metabolismo , Animales , Antígenos CD/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Concanavalina A/administración & dosificación , Concanavalina A/toxicidad , Citocinas/metabolismo , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Galactosilceramidas/administración & dosificación , Galactosilceramidas/toxicidad , Hígado/efectos de los fármacos , Hígado/inmunología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Células T Asesinas Naturales/inmunología , Receptores Inmunológicos/genética , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Análisis de Supervivencia
13.
Stem Cell Reports ; 8(6): 1630-1644, 2017 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-28552607

RESUMEN

Bone marrow-derived mesenchymal stem cells (BMSCs) are proposed as the cells of origin of several subtypes of osteosarcoma (OS). However, signals that direct BMSCs to form different subtypes of OS are unclear. Here we show that the default tumor type from spontaneously transformed p53 knockout (p53_KO) BMSCs is osteoblastic OS. The development of this default tumor type caused by p53 loss can be overridden by various oncogenic signals: RAS reprograms p53_KO BMSCs into undifferentiated sarcoma, AKT enhances osteoblastic OS, while cFOS promotes chondroblastic OS formation. We focus on studying the mechanism of cFOS-induced chondroblastic OS formation. Integrated genome-wide studies reveal a regulatory mechanism whereby cFOS binds to the promoter of a key chondroblastic transcription factor, Sox9, and induces its transcription in BMSCs. Importantly, SOX9 mediates cFOS-induced cartilage formation in chondroblastic OS. In summary, oncogenes determine tumor types derived from BMSCs, and the cFOS-SOX9 axis is critical for chondroblastic OS formation.


Asunto(s)
Células de la Médula Ósea/citología , Neoplasias Óseas/patología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Osteosarcoma/patología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Factor de Transcripción SOX9/metabolismo , Animales , Neoplasias Óseas/metabolismo , Neoplasias Óseas/terapia , Diferenciación Celular , Reprogramación Celular , Condrocitos/citología , Condrocitos/metabolismo , Condrogénesis , Humanos , Células Madre Mesenquimatosas/citología , Ratones , Ratones Noqueados , Osteogénesis , Osteosarcoma/metabolismo , Osteosarcoma/terapia , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , Factor de Transcripción SOX9/antagonistas & inhibidores , Factor de Transcripción SOX9/genética , Transducción de Señal , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas ras/metabolismo
14.
Exp Mol Med ; 34(6): 401-10, 2002 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-12526081

RESUMEN

Rat hippocampal precursor cells isolated from hippocampi of embryonic day 16.5 (E16.5) rat embryos were found to proliferate in the presence of basic fibroblast growth factor. Addition of soluble neural cell adhesion molecule (NCAM) to these precursor cells reduced cell proliferation in a dose dependent manner and enhanced the induction of precursor cells' differentiation to the neuronal lineage. Given these findings that NCAM induces the differentiation of hippocampal precursor cells, we investigated possible effects of NCAM on the expression of basic helix-loop-helix (bHLH) transcription factors during the differentiation. Soluble NCAM upregulated the transcription of bHLH transcription factors, neurogenin1 and NeuroD, but decreased HES5. Western blot analysis showed that NCAM increased the expression levels of CaMKII, p-MAPK, GluR1 and NR1 but decreased p-STAT3. These results support a role for NCAM in the inhibition of proliferation and the induction of neural differentiation of hippocampal neural precursor cells, and act as developmental regulators of the bHLH families, ultimately leading to the generation of glutamatergic neural cell types in the differentiation of hippocampal precursor cells.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Hipocampo/citología , Hipocampo/efectos de los fármacos , Moléculas de Adhesión de Célula Nerviosa/farmacología , Neuronas/efectos de los fármacos , Receptores de Glutamato/metabolismo , Células Madre/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , División Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Células Cultivadas , Secuencias Hélice-Asa-Hélice , Neuronas/citología , Neuronas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Transducción de Señal , Células Madre/citología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
15.
Cell Biosci ; 3(1): 42, 2013 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-24171803

RESUMEN

The viability and subtle developmental defects of p53 knockout mice suggest that p53 does not play major role in development. However, contradictory evidence also exists. This discrepancy mainly results from the lack of molecular and cellular mechanisms and the general fact that p53 activation requires stresses. Recent studies of p53 in mouse and human ES cells and induced pluripotent stem (iPS) cells shed new light on the mechanisms of the developmental roles of p53. This review summarizes these new studies that support the developmental roles of p53, highlights the possible underlying molecular mechanisms, and discusses the potential relationship between the developmental roles and the tumor suppressive function of p53. In summary, the molecular mechanisms underlying the developmental roles of p53 are emerging, and the developmental roles and tumor suppressive function of p53 may be closely related.

16.
Cell Cycle ; 11(9): 1796-803, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22453082

RESUMEN

Upon genotoxic stress and during normal S phase, ATM phosphorylates the checkpoint clamp protein Rad9 in a manner that depends on Ser272. Ser272 is the only known ATM-dependent phosphorylation site in human Rad9. However, Ser272 phosphorylation is not required for survival or checkpoint activation after DNA damage. The physiological function of Ser272 remains elusive. Here, we show that ATM-dependent Rad9(Ser272) phosphorylation requires the MRN complex and controls repair pathways. Furthermore, the mutant cells accumulate large numbers of chromosome breaks and induce gross chromosomal rearrangements. Our findings establish a new and unexpected role for ATM: it phosphorylates the checkpoint clamp in order to control repair pathways, thereby maintaining genomic integrity during unperturbed cell cycle and upon DNA damage.


Asunto(s)
Puntos de Control del Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Inestabilidad Genómica , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ácido Anhídrido Hidrolasas , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Rotura Cromosómica , Roturas del ADN de Doble Cadena , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Células HeLa , Humanos , Proteína Homóloga de MRE11 , Morfolinas/farmacología , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Mutagénesis Sitio-Dirigida , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oxidación-Reducción , Fosforilación , Plásmidos/genética , Plásmidos/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Pironas/farmacología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Reparación del ADN por Recombinación , Serina/metabolismo , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/genética
17.
Mol Cell Biol ; 29(18): 4878-90, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19564421

RESUMEN

In noradrenergic progenitors, Phox2a mediates cell cycle exit and neuronal differentiation by inducing p27(Kip1) transcription in response to activation of the cyclic AMP (cAMP) pathway. The mechanism of cAMP-mediated activation of Phox2a is unknown. We identified a cluster of phosphoserine-proline sites in Phox2a by mass spectrometry. Ser206 appeared to be the most prominent phosphorylation site. A phospho-Ser206 Phox2a antibody detected dephosphorylation of Phox2a that was dependent on activation of the cAMP pathway, which occurred prior to neuronal differentiation of noradrenergic CAD cells. Employing serine-to-alanine and serine-to-aspartic acid Phox2a substitution mutants expressed in inducible CAD cell lines, we demonstrated that the transcriptional activity of Phox2a is regulated by two sequential cAMP-dependent events: first, cAMP signaling promotes dephosphorylation of Phox2a in at least one site, Ser206, thereby allowing Phox2a to bind DNA and initiate p27(Kip1) transcription; second, following dephosphorylation of the phosphoserine cluster (Ser202 and Ser208), Phox2a becomes phosphorylated by protein kinase A (PKA) on Ser153, which prevents association of Phox2a with DNA and terminates p27(Kip1) transcription. This represents a novel mechanism by which the same stimulus, cAMP signaling, first activates Phox2a by dephosphorylation of Ser206 and then, after a built-in delay, inactivates Phox2a via PKA-dependent phosphorylation of Ser153, thereby modulating onset and duration of p27(Kip1) transcription.


Asunto(s)
AMP Cíclico/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Proteínas de Homeodominio/metabolismo , Transducción de Señal , Transcripción Genética , Alanina/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Diferenciación Celular , Línea Celular , Inmunoprecipitación de Cromatina , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , ADN/metabolismo , Regulación de la Expresión Génica , Proteínas de Homeodominio/química , Proteínas de Homeodominio/genética , Proteínas de Filamentos Intermediarios/metabolismo , Espectrometría de Masas , Glicoproteínas de Membrana/metabolismo , Modelos Biológicos , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Neuronas/metabolismo , Periferinas , Fosforilación , Fosfoserina/metabolismo , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA