Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Int J Mol Sci ; 25(8)2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38674001

RESUMEN

Medulloblastoma (MB) encompasses diverse subgroups, and leptomeningeal disease/metastasis (LMD) plays a substantial role in associated fatalities. Despite extensive exploration of canonical genes in MB, the molecular mechanisms underlying LMD and the involvement of the orthodenticle homeobox 2 (OTX2) gene, a key driver in aggressive MB Group 3, remain insufficiently understood. Recognizing OTX2's pivotal role, we investigated its potential as a catalyst for aggressive cellular behaviors, including migration, invasion, and metastasis. OTX2 overexpression heightened cell growth, motility, and polarization in Group 3 MB cells. Orthotopic implantation of OTX2-overexpressing cells in mice led to reduced median survival, accompanied by the development of spinal cord and brain metastases. Mechanistically, OTX2 acted as a transcriptional activator of the Mechanistic Target of Rapamycin (mTOR) gene's promoter and the mTORC2 signaling pathway, correlating with upregulated downstream genes that orchestrate cell motility and migration. Knockdown of mTOR mRNA mitigated OTX2-mediated enhancements in cell motility and polarization. Analysis of human MB tumor samples (N = 952) revealed a positive correlation between OTX2 and mTOR mRNA expression, emphasizing the clinical significance of OTX2's role in the mTORC2 pathway. Our results reveal that OTX2 governs the mTORC2 signaling pathway, instigating LMD in Group 3 MBs and offering insights into potential therapeutic avenues through mTORC2 inhibition.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Diana Mecanicista del Complejo 2 de la Rapamicina , Meduloblastoma , Neoplasias Meníngeas , Factores de Transcripción Otx , Animales , Femenino , Humanos , Masculino , Ratones , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/patología , Neoplasias Cerebelosas/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/genética , Meduloblastoma/genética , Meduloblastoma/patología , Meduloblastoma/metabolismo , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/patología , Neoplasias Meníngeas/metabolismo , Neoplasias Meníngeas/secundario , Factores de Transcripción Otx/metabolismo , Factores de Transcripción Otx/genética , Transducción de Señal
2.
Pediatr Blood Cancer ; 64(12)2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28598542

RESUMEN

BACKGROUND: Overexpression of minichromosome maintenance (MCM) proteins 2, 3, and 7 is associated with migration and invasion in medulloblastoma (MB). However, expression profiling of all prereplication complex (pre-RC) has not been addressed in MBs. PROCEDURE: We performed mRNA expression profiling of a large set of pre-RC elements in cell lines and tumor tissues of MB. RNAi technology was employed for functional studies in MB cell lines. RESULTS: Our data showed that most of the pre-RC components are significantly overexpressed in MB. Among all pre-RC mRNAs, MCM10 showed the highest level of expression (∼500- to 1,000-fold) in MB cell lines and tissues compared to the levels detected in cerebellum. In addition, RNAi silencing of MCM10 caused reduced cell proliferation and cell viability in MB cells. CONCLUSIONS: Taken together, our study reveals that the pre-RC is dysregulated in MB. In addition, MCM10, a member of this complex, is significantly overexpressed in MB and is required for tumor cell proliferation.


Asunto(s)
Neoplasias Cerebelosas/química , Meduloblastoma/química , Proteínas de Mantenimiento de Minicromosoma/fisiología , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Neoplasias Cerebelosas/patología , Humanos , Inmunohistoquímica , Meduloblastoma/patología , Proteínas de Mantenimiento de Minicromosoma/análisis
3.
Proc Natl Acad Sci U S A ; 110(44): 17844-9, 2013 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-24127576

RESUMEN

The major mammalian apurinic/apyrimidinic endonuclease Ape1 is a multifunctional protein operating in protection of cells from oxidative stress via its DNA repair, redox, and transcription regulatory activities. The importance of Ape1 has been marked by previous work demonstrating its requirement for viability in mammalian cells. However, beyond a requirement for Ape1-dependent DNA repair activity, deeper molecular mechanisms of the fundamental role of Ape1 in cell survival have not been defined. Here, we report that Ape1 is an essential factor stabilizing telomeric DNA, and its deficiency is associated with telomere dysfunction and segregation defects in immortalized cells maintaining telomeres by either the alternative lengthening of telomeres pathway (U2OS) or telomerase expression (BJ-hTERT), or in normal human fibroblasts (IMR90). Through the expression of Ape1 derivatives with site-specific changes, we found that the DNA repair and N-terminal acetylation domains are required for the Ape1 function at telomeres. Ape1 associates with telomere proteins in U2OS cells, and Ape1 depletion causes dissociation of TRF2 protein from telomeres. Consistent with this effect, we also observed that Ape1 depletion caused telomere shortening in both BJ-hTERT and in HeLa cells. Thus, our study describes a unique and unpredicted role for Ape1 in telomere protection, providing a direct link between base excision DNA repair activities and telomere metabolism.


Asunto(s)
Reparación del ADN/fisiología , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Homeostasis del Telómero/genética , Western Blotting , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Cartilla de ADN/genética , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Hibridación Fluorescente in Situ , Telomerasa/metabolismo , Homeostasis del Telómero/fisiología
4.
Cancers (Basel) ; 15(15)2023 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-37568598

RESUMEN

Glioblastoma is one of the most devastating neoplasms of the central nervous system. This study focused on the development of serum extracellular vesicle (EV)-based glioblastoma tumor marker panels that can be used in a clinic to diagnose glioblastomas and to monitor tumor burden, progression, and regression in response to treatment. RNA sequencing studies were performed using RNA isolated from serum EVs from both patients (n = 85) and control donors (n = 31). RNA sequencing results for preoperative glioblastoma EVs compared to control EVs revealed 569 differentially expressed genes (DEGs, 2XFC, FDR < 0.05). By using these DEGs, we developed serum-EV-based biomarker panels for the following glioblastomas: wild-type IDH1 (96% sensitivity/80% specificity), MGMT promoter methylation (91% sensitivity/73% specificity), p53 gene mutation (100% sensitivity/89% specificity), and TERT promoter mutation (89% sensitivity/100% specificity). This is the first study showing that serum-EV-based biomarker panels can be used to diagnose glioblastomas with a high sensitivity and specificity.

5.
Cancers (Basel) ; 14(8)2022 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-35454889

RESUMEN

Extracellular vesicles (EVs) may be used as a non-invasive screening platform to discover markers associated with early diagnosis, prognosis, and treatment response. Such an approach is invaluable for diseases such as glioblastoma, for which only a few non-invasive diagnostic or prognostic markers are available. We used mass spectrometry to analyze proteomics profiles of EVs derived from four glioblastoma cell lines and human primary astrocytes (HPAs) and found that SRPX is the only protein enriched in the majority of glioblastoma EVs that was absent in the HPA-derived EVs. Then, we evaluated the relationship between SRPX protein expression and tumor grade using immunohistochemical staining (IHC) and performed colony formation and viability assays to analyze the possible function of SRPX in glioblastoma. SRPX mRNA and protein expression were associated with tumor grade. Moreover, temozolomide (TMZ)-resistant tumor tissues showed highly positive SRPX staining, compared to all other tumor grades. Additionally, glioblastoma cells displayed enhanced SRPX gene expression when exposed to TMZ. Knockdown of SRPX gene expression via siRNA inhibited cell viability. Taken together, the results of this study suggest that SRPX can be used as a novel tumor marker for diagnostic and prognostic purposes and can also be a therapeutic target for glioblastomas.

6.
Front Cell Dev Biol ; 9: 651818, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33855027

RESUMEN

Cancer metastasis is the major cause of death from cancer (Massague and Obenauf, 2016; Steeg, 2016). The extensive genetic heterogeneity and cellular plasticity of metastatic tumors set a prime barrier for the current cancer treatment protocols (Boumahdi and de Sauvage, 2020). In addition, acquired therapy resistance has become an insurmountable obstacle that abolishes the beneficial effects of numerous anti-cancer regimens (De Angelis et al., 2019; Boumahdi and de Sauvage, 2020). Here we report that deficiency of Ku leads to the exploitation of host cells in human cancer cell line models. We found that, upon conditional deletion of XRCC6 that codes for Ku70, HCT116 human colorectal cancer cells gain a parasitic lifestyle that is characterized by the continuous cycle of host cell exploitation. We also found that DAOY cells, a human medulloblastoma cell line, innately lack nuclear Ku70/Ku86 proteins and utilize the host-cell invasion/exit mechanism for maintenance of their survival, similarly to the Ku70 conditionally-null HCT116 cells. Our study demonstrates that a functional loss of Ku protein promotes an adaptive, opportunistic switch to a parasitic lifestyle in human cancer cells, providing evidence for a previously unknown mechanism of cell survival in response to severe genomic stress. We anticipate that our study will bring a new perspective for understanding the mechanisms of cancer cell evolution, leading to a shift in the current concepts of cancer therapy protocols directed to the prevention of cancer metastasis and therapy resistance.

7.
Nucleic Acids Res ; 35(17): 5706-16, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17715146

RESUMEN

Werner syndrome (WS) is a severe recessive disorder characterized by premature aging, cancer predisposition and genomic instability. The gene mutated in WS encodes a bi-functional enzyme called WRN that acts as a RecQ-type DNA helicase and a 3'-5' exonuclease, but its exact role in DNA metabolism is poorly understood. Here we show that WRN physically interacts with the MSH2/MSH6 (MutSalpha), MSH2/MSH3 (MutSbeta) and MLH1/PMS2 (MutLalpha) heterodimers that are involved in the initiation of mismatch repair (MMR) and the rejection of homeologous recombination. MutSalpha and MutSbeta can strongly stimulate the helicase activity of WRN specifically on forked DNA structures with a 3'-single-stranded arm. The stimulatory effect of MutSalpha on WRN-mediated unwinding is enhanced by a G/T mismatch in the DNA duplex ahead of the fork. The MutLalpha protein known to bind to the MutS alpha-heteroduplex complexes has no effect on WRN-mediated DNA unwinding stimulated by MutSalpha, nor does it affect DNA unwinding by WRN alone. Our data are consistent with results of genetic experiments in yeast suggesting that MMR factors act in conjunction with a RecQ-type helicase to reject recombination between divergent sequences.


Asunto(s)
Disparidad de Par Base , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , RecQ Helicasas/metabolismo , Sitios de Unión , Línea Celular , ADN/química , ADN/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Exodesoxirribonucleasas , Humanos , Proteínas MutL , Proteína 2 Homóloga a MutS/metabolismo , Proteína 3 Homóloga de MutS , Estructura Terciaria de Proteína , RecQ Helicasas/química , Técnicas del Sistema de Dos Híbridos , Helicasa del Síndrome de Werner
8.
Methods Mol Biol ; 1895: 87-96, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30539531

RESUMEN

Gene therapy involves the introduction of genes (termed transgenes) into cells to compensate for a deficiency or to make a beneficial protein. Gene therapy can used as a form of cancer treatment. A particularly attractive paradigm in this regard involves the selective introduction of transgenes into cancer cells that converts inactive prodrugs into active chemotherapeutic agents, thereby triggering the death of cancer cells. Since prodrugs are inactive, they tend not to cause significant side-effects and are well-tolerated by patients relative to conventional chemotherapy. Several viral and nonviral vectors have been used as delivery tools for suicide gene therapy. Extracellular vesicles (EVs) are now recognized as a promising class of nonviral delivery vectors. Here, we describe a method in which a suicide fusion gene construct is loaded into EVs derived from a non-tumorigenic cell line. Delivery of these modified EVs to glioblastoma cell lines and spheroids decreases glioblastoma cell viability, induces apoptotic cell death, and inhibits tumor growth in vivo.


Asunto(s)
Portadores de Fármacos , Vesículas Extracelulares , Genes Transgénicos Suicidas , Terapia Genética/métodos , Glioblastoma/terapia , Línea Celular Tumoral , Citosina Desaminasa/metabolismo , Proteínas Fúngicas/metabolismo , Glioblastoma/tratamiento farmacológico , Células HEK293 , Humanos , Pentosiltransferasa/metabolismo , Profármacos/metabolismo , Profármacos/uso terapéutico , ARN Mensajero , Levaduras/enzimología
9.
Front Oncol ; 9: 1031, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31649887

RESUMEN

Meningiomas are primary central nervous system (CNS) tumors that originate from the arachnoid cells of the meninges. Recurrence occurs in higher grade meningiomas and a small subset of Grade I meningiomas with benign histology. Currently, there are no established circulating tumor markers which can be used for diagnostic and prognostic purposes in a non-invasive way for meningiomas. Here, we aimed to identify potential biomarkers of meningioma in patient sera. For this purpose, we collected preoperative (n = 30) serum samples from the meningioma patients classified as Grade I (n = 23), Grade II (n = 4), or Grade III (n = 3). We used a high-throughput, multiplex immunoassay cancer panel comprising of 92 cancer-related protein biomarkers to explore the serum protein profiles of meningioma patients. We detected 14 differentially expressed proteins in the sera of the Grade I meningioma patients in comparison to the age- and gender-matched control subjects (n = 12). Compared to the control group, Grade I meningioma patients showed increased serum levels of amphiregulin (AREG), CCL24, CD69, prolactin, EGF, HB-EGF, caspase-3, and decreased levels of VEGFD, TGF-α, E-Selectin, BAFF, IL-12, CCL9, and GH. For validation studies, we utilized an independent set of meningioma tumor tissue samples (Grade I, n = 20; Grade II, n = 10; Grade III, n = 6), and found that the expressions of amphiregulin and Caspase3 are significantly increased in all grades of meningiomas either at the transcriptional or protein level, respectively. In contrast, the gene expression of VEGF-D was significantly lower in Grade I meningioma tissue samples. Taken together, our study identifies a meningioma-specific protein signature in blood circulation of meningioma patients and highlights the importance of equilibrium between tumor-promoting factors and anti-tumor immunity.

10.
Nucleic Acids Res ; 34(18): 5217-31, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17003056

RESUMEN

The role of the human RECQ5beta helicase in the maintenance of genomic stability remains elusive. Here we show that RECQ5beta promotes strand exchange between arms of synthetic forked DNA structures resembling a stalled replication fork in a reaction dependent on ATP hydrolysis. BLM and WRN can also promote strand exchange on these structures. However, in the presence of human replication protein A (hRPA), the action of these RecQ-type helicases is strongly biased towards unwinding of the parental duplex, an effect not seen with RECQ5beta. A domain within the non-conserved portion of RECQ5beta is identified as being important for its ability to unwind the lagging-strand arm and to promote strand exchange on hRPA-coated forked structures. We also show that RECQ5beta associates with DNA replication factories in S phase nuclei and persists at the sites of stalled replication forks after exposure of cells to UV irradiation. Moreover, RECQ5beta is found to physically interact with the polymerase processivity factor proliferating cell nuclear antigen in vitro and in vivo. Collectively, these findings suggest that RECQ5beta may promote regression of stalled replication forks to facilitate the bypass of replication-blocking lesions by template-switching. Loss of such activity could explain the elevated level of mitotic crossovers observed in RECQ5beta-deficient cells.


Asunto(s)
Replicación del ADN , ADN/química , RecQ Helicasas/metabolismo , Adenosina Trifosfatasas/metabolismo , Línea Celular , Daño del ADN , ADN Helicasas/metabolismo , Células HeLa , Humanos , Oligonucleótidos/química , Antígeno Nuclear de Célula en Proliferación/metabolismo , Estructura Terciaria de Proteína , RecQ Helicasas/química , Proteína de Replicación A/metabolismo , Moldes Genéticos
11.
Sci Rep ; 7(1): 9674, 2017 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-28852018

RESUMEN

Ape1 is the major apurinic/apyrimidinic (AP) endonuclease activity in mammalian cells, and a key factor in base-excision repair of DNA. High expression or aberrant subcellular distribution of Ape1 has been detected in many cancer types, correlated with drug response, tumor prognosis, or patient survival. Here we present evidence that Ape1 facilitates BRCA1-mediated homologous recombination repair (HR), while counteracting error-prone non-homologous end joining of DNA double-strand breaks. Furthermore, Ape1, coordinated with checkpoint kinase Chk2, regulates drug response of glioblastoma cells. Suppression of Ape1/Chk2 signaling in glioblastoma cells facilitates alternative means of damage site recruitment of HR proteins as part of a genomic defense system. Through targeting "HR-addicted" temozolomide-resistant glioblastoma cells via a chemical inhibitor of Rad51, we demonstrated that targeting HR is a promising strategy for glioblastoma therapy. Our study uncovers a critical role for Ape1 in DNA repair pathway choice, and provides a mechanistic understanding of DNA repair-supported chemoresistance in glioblastoma cells.


Asunto(s)
Reparación del ADN , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Tolerancia a Medicamentos , Glioblastoma/patología , Redes y Vías Metabólicas , Ubiquitina-Proteína Ligasas/metabolismo , Quinasa de Punto de Control 2/metabolismo , Recombinación Homóloga , Humanos
12.
Environ Health Perspect ; 110 Suppl 5: 813-7, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12426137

RESUMEN

Metallothioneins are small, cysteine-rich, metal-binding proteins that play important roles in maintaining intracellular metal homeostasis and in transition metal detoxification. MTF-1 (metal transcription factor-1) plays a central role in regulating the metal-inducible, transcriptional activation of metallothionein. Here we report that the phosphorylation of MTF-1 plays a critical role in the activation of MTF-1/metal-responsive element-mediated transcription. Inhibitor studies indicate that signal transduction cascades, including those mediated by protein kinase C, tyrosine kinase, and casein kinase II, are essential for zinc- and cadmium-inducible transcription. In addition, calcium signaling is also involved in regulating transcription. In contrast, cAMP-dependent protein kinase may not be directly involved in the metal response. Contrary to what has been reported for other transcription factors, the inhibition of transcriptional activation does not impair the binding of MTF-1 to DNA, suggesting that phosphorylation is not regulating DNA binding. Elevated phosphorylation of MTF-1 is observed under conditions of protein kinase C inhibition, suggesting that dephosphorylation of this transcription factor mediates its activation.


Asunto(s)
Metalotioneína/metabolismo , Metales Pesados/farmacología , Factores de Transcripción/farmacología , Animales , Células COS , Proteínas Quinasas Dependientes de AMP Cíclico/farmacología , Aductos de ADN , Proteínas de Unión al ADN , Células HeLa , Homeostasis , Humanos , Metales Pesados/efectos adversos , Fosforilación , Plásmidos , Proteínas Quinasas , Transducción de Señal , Transcripción Genética/efectos de los fármacos , Factor de Transcripción MTF-1
13.
Cancer Res ; 71(3): 852-61, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21156648

RESUMEN

Micro RNAs (miRNA) negatively regulate protein-coding genes at the posttranscriptional level and are critical in tumorigenesis. Schwannomas develop from proliferation of dedifferentiated Schwann cells, which normally wrap nerve fibers to help support and insulate nerves. In this study, we carried out high-throughput miRNA expression profiling of human vestibular schwannomas by using an array representing 407 known miRNAs to explore the role of miRNAs in tumor growth. Twelve miRNAs were found to be significantly deregulated in tumor samples as compared with control nerve tissue, defining a schwannoma-typical signature. Among these miRNAs, we focused on miR-7, which was one of the most downregulated in these tumors and has several known oncogene targets, including mRNAs for epidermal growth factor receptor (EGFR) and p21-activated kinase 1 (Pak1). We found that overexpression of miR-7 inhibited schwannoma cell growth both in culture and in xenograft tumor models in vivo, which correlated with downregulation of these signaling pathways. Furthermore, we identified a novel direct target of miR-7, the mRNA for associated cdc42 kinase 1 (Ack1), with the expression levels of miR-7 and Ack1 being inversely correlated in human schwannoma samples. These results represent the first miRNA profiling of schwannomas and the first report of a tumor suppressor function for miR-7 in these tumors that is mediated by targeting the EGFR, Pak1, and Ack1 oncogenes. Our findings suggest miR-7 as a potential therapeutic molecule for schwannoma treatment, and they prompt clinical evaluation of drugs that can inhibit the EGFR, Pak1, and Ack1 signaling pathways to treat this tumor type.


Asunto(s)
MicroARNs/genética , Neurilemoma/genética , Neurilemoma/metabolismo , Animales , Procesos de Crecimiento Celular/genética , Línea Celular Tumoral , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/biosíntesis , Receptores ErbB/metabolismo , Femenino , Humanos , Ratones , Ratones Desnudos , MicroARNs/biosíntesis , Neurilemoma/patología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/biosíntesis , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal , Transfección , Trasplante Heterólogo , Regulación hacia Arriba , Quinasas p21 Activadas/antagonistas & inhibidores , Quinasas p21 Activadas/biosíntesis , Quinasas p21 Activadas/metabolismo
14.
J Biol Chem ; 278(34): 31879-83, 2003 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-12805380

RESUMEN

Mammalian cells react to heavy metal stress by transcribing a number of genes that contain metal-response elements (MREs) in their promoter/enhancer region; this activation is mediated by metal-responsive transcription factor-1 (MTF-1). Well-known target genes of MTF-1 are those encoding metallothioneins, small, cysteine-rich proteins with a high affinity for heavy metals. The response to heat shock, another cell stress, is mediated by heat shock transcription factor 1 (HSF1), which activates a battery of heat shock genes. Little is known about the cross-talk between the different anti-stress systems of the cell. Here we report a synergistic activation of metal-responsive promoters by heavy metal load (zinc or cadmium) and heat shock. An obvious explanation, cooperativity between MTF-1 and HSF1, seems unlikely: transfected HSF1 boosts the activity of an Hsp70 promoter but hardly affects an MRE-containing promoter upon exposure to metal and heat shock. A clue to the mechanism is given by our finding that heat shock leads to intracellular accumulation of heavy metals. We propose that the known anti-apoptotic effect of heat shock proteins allows for cell survival despite heavy metal accumulation and, consequently, results in a hyperactivation of the metal response pathway.


Asunto(s)
Calor , Metales Pesados/farmacología , Factores de Transcripción/fisiología , Transcripción Genética , Línea Celular , Proteínas de Unión al ADN , Humanos , Regiones Promotoras Genéticas , Transporte de Proteínas , Factores de Transcripción/metabolismo , Transcripción Genética/efectos de los fármacos , Factor de Transcripción MTF-1
15.
J Biol Chem ; 277(23): 20438-45, 2002 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-11923282

RESUMEN

Every living organism must detoxify nonessential metals and carefully control the intracellular concentration of essential metals. Metallothioneins, which are small, cysteine-rich, metal-binding proteins, play an important role in these processes. In addition, the transcription of their cognate genes is activated in response to metal exposure. The zinc finger transcription factor MTF-1 plays a central role in the metal-inducible transcriptional activation of metallothionein and other genes involved in metal homeostasis and cellular stress response. Here we report that the phosphorylation of MTF-1 plays a critical role in its activation by zinc and cadmium. Inhibitor studies indicate that multiple kinases and signal transduction cascades, including those mediated by protein kinase C, tyrosine kinase, and casein kinase II, are essential for zinc- and cadmium-inducible transcriptional activation. In addition, calcium signaling is also involved in regulating metal-activated transcription. In contrast, cAMP-dependent protein kinase may not be directly involved in the metal response. Contrary to what has been reported for other transcription factors, inhibition of transcriptional activation does not impair the binding of MTF-1 to DNA, suggesting that phosphorylation is not regulating DNA binding. Elevated phosphorylation of MTF-1 is observed under condition of protein kinase C inhibition, suggesting that specific dephosphorylation of this transcription factor contributes to its activation.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Metalotioneína/genética , Transducción de Señal/fisiología , Factores de Transcripción/fisiología , Transcripción Genética/fisiología , Animales , Línea Celular , Cloranfenicol O-Acetiltransferasa/genética , Proteínas de Unión al ADN , Ensayo de Cambio de Movilidad Electroforética , Ensayo de Inmunoadsorción Enzimática , Humanos , Fosforilación , Factor de Transcripción MTF-1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA