Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 189
Filtrar
1.
Antiviral Res ; 225: 105870, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38556059

RESUMEN

Following acute human alphaherpesvirus 1 (HSV-1) infection of oral-facial mucosal surfaces, sensory neurons in trigeminal ganglia (TG) are important sites for life-long latency. Neurons in the central nervous system, including brainstem, also harbor viral genomes during latency. Periodically, certain cellular stressors trigger reactivation from latency, which can lead to recurrent HSV-1 disease: herpes labialis, herpes stromal keratitis, and encephalitis for example. Activation of the glucocorticoid receptor (GR) by stressful stimuli enhances HSV-1 gene expression, replication, and explant-induced reactivation. GR and certain stress-induced Krüppel like factors (KLF) cooperatively transactivate cis-regulatory modules (CRM) that drive expression of viral transcriptional regulatory proteins (ICP0, ICP4, and ICP27). These CRMs lack GR response elements (GRE); however, specificity protein 1 (Sp1) binding sites are crucial for GR and KLF15 or KLF4 mediated transactivation. Hence, we tested whether Sp1 or Sp3 regulate viral replication and transactivation of the ICP0 promoter. During early stages of explant-induced reactivation from latency, the number of Sp3+ TG neurons were significantly higher relative to TG from latently infected mice. Conversely, Sp1+ TG neurons were only increased in females, but not male mice, during explant-induced reactivation. Sp1 siRNA significantly reduced HSV-1 replication in cultured mouse (Neuro-2A) and monkey (CV-1) cells. Mithramycin A, an antibiotic that has anti-tumor activity preferentially interacts with GC-rich DNA, including Sp1 binding sites, significantly reduced HSV-1 replication indicating it has antiviral activity. GR and Sp1 or Sp3 transactivated the HSV-1 ICP0 promoter in Neuro-2A and CV-1 cells confirming these transcription factors enhance viral replication and gene expression.


Asunto(s)
Herpes Simple , Herpesvirus Humano 1 , Proteínas Inmediatas-Precoces , Plicamicina/análogos & derivados , Femenino , Humanos , Ratones , Animales , Herpesvirus Humano 1/genética , Receptores de Glucocorticoides/metabolismo , Activación Viral , Latencia del Virus/genética , Proteínas Inmediatas-Precoces/genética , Antibacterianos , Ubiquitina-Proteína Ligasas/genética
2.
J Virol ; 98(1): e0143623, 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38084958

RESUMEN

Bovine alphaherpesvirus 1 (BoHV-1) infections cause respiratory tract disorders and suppress immune responses, which can culminate in bacterial pneumonia. Following acute infection, BoHV-1 establishes lifelong latency in sensory neurons present in trigeminal ganglia (TG) and unknown cells in pharyngeal tonsil. Latently infected calves consistently reactivate from latency after an intravenous injection of the synthetic corticosteroid dexamethasone (DEX), which mimics the effects of stress. The immediate early transcription unit 1 (IEtu1) promoter drives expression of infected cell protein 0 (bICP0) and bICP4, two key viral transcriptional regulators. The IEtu1 promoter contains two functional glucocorticoid receptor (GR) response elements (GREs), and this promoter is transactivated by GR, DEX, and certain Krüppel transcription factors that interact with GC-rich motifs, including consensus specificity protein 1 (Sp1) binding sites. Based on these observations, we hypothesized that Sp1 stimulates productive infection and transactivates key BoHV-1 promoters. DEX treatment of latently infected calves increased the number of Sp1+ TG neurons and cells in pharyngeal tonsil indicating that Sp1 expression is induced by stress. Silencing Sp1 protein expression with siRNA or mithramycin A, a drug that preferentially binds GC-rich DNA, significantly reduced BoHV-1 replication. Moreover, BoHV-1 infection of permissive cells increased Sp1 steady-state protein levels. In transient transfection studies, GR and Sp1 cooperatively transactivate IEtu1 promoter activity unless both GREs are mutated. Co-immunoprecipitation studies revealed that GR and Sp1 interact in mouse neuroblastoma cells (Neuro-2A) suggesting this interaction stimulates IEtu1 promoter activity. Collectively, these studies suggested that the cellular transcription factor Sp1 enhances productive infection and stress-induced BoHV-1 reactivation from latency.IMPORTANCEFollowing acute infection, bovine alphaherpesvirus 1 (BoHV-1) establishes lifelong latency in sensory neurons in trigeminal ganglia (TG) and pharyngeal tonsil. The synthetic corticosteroid dexamethasone consistently induces BoHV-1 reactivation from latency. The number of TG neurons and cells in pharyngeal tonsil expressing the cellular transcription factor specificity protein 1 (Sp1) protein increases during early stages of dexamethasone-induced reactivation from latency. Silencing Sp1 expression impairs BoHV-1 replication in permissive cells. Interestingly, mithramycin A, a neuroprotective antibiotic that preferentially binds GC-rich DNA, impairs Sp1 functions and reduces BoHV-1 replication suggesting that it is a potential antiviral drug. The glucocorticoid receptor (GR) and Sp1 cooperatively transactivate the BoHV-1 immediate early transcript unit 1 (IEtu1) promoter, which drives expression of infected cell protein 0 (bICP0) and bICP4. Mithramycin A also reduced Sp1- and GR-mediated transactivation of the IEtu1 promoter. These studies revealed that GR and Sp1 trigger viral gene expression and replication following stressful stimuli.


Asunto(s)
Infecciones por Herpesviridae , Herpesvirus Bovino 1 , Receptores de Glucocorticoides , Factor de Transcripción Sp1 , Animales , Bovinos , Ratones , Corticoesteroides/metabolismo , Dexametasona/farmacología , ADN/metabolismo , Herpesvirus Bovino 1/fisiología , Plicamicina/análogos & derivados , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Factores de Transcripción/metabolismo , Proteínas Virales/metabolismo , Factor de Transcripción Sp1/metabolismo
3.
J Nanobiotechnology ; 19(1): 267, 2021 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-34488783

RESUMEN

BACKGROUND: Sarcomas comprise a group of aggressive malignancies with very little treatment options beyond standard chemotherapy. Reposition of approved drugs represents an attractive approach to identify effective therapeutic compounds. One example is mithramycin (MTM), a natural antibiotic which has demonstrated a strong antitumour activity in several tumour types, including sarcomas. However, its widespread use in the clinic was limited by its poor toxicity profile. RESULTS: In order to improve the therapeutic index of MTM, we have loaded MTM into newly developed nanocarrier formulations. First, polylactide (PLA) polymeric nanoparticles (NPs) were generated by nanoprecipitation. Also, liposomes (LIP) were prepared by ethanol injection and evaporation solvent method. Finally, MTM-loaded hydrogels (HG) were obtained by passive loading using a urea derivative non-peptidic hydrogelator. MTM-loaded NPs and LIP display optimal hydrodynamic radii between 80 and 105 nm with a very low polydispersity index (PdI) and encapsulation efficiencies (EE) of 92 and 30%, respectively. All formulations show a high stability and different release rates ranging from a fast release in HG (100% after 30 min) to more sustained release from NPs (100% after 24 h) and LIP (40% after 48 h). In vitro assays confirmed that all assayed MTM formulations retain the cytotoxic, anti-invasive and anti-stemness potential of free MTM in models of myxoid liposarcoma, undifferentiated pleomorphic sarcoma and chondrosarcoma. In addition, whole genome transcriptomic analysis evidenced the ability of MTM, both free and encapsulated, to act as a multi-repressor of several tumour-promoting pathways at once. Importantly, the treatment of mice bearing sarcoma xenografts showed that encapsulated MTM exhibited enhanced therapeutic effects and was better tolerated than free MTM. CONCLUSIONS: Overall, these novel formulations may represent an efficient and safer MTM-delivering alternative for sarcoma treatment.


Asunto(s)
Plicamicina/análogos & derivados , Plicamicina/farmacología , Plicamicina/uso terapéutico , Sarcoma/patología , Animales , Antibacterianos/uso terapéutico , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Condrosarcoma/tratamiento farmacológico , Composición de Medicamentos , Femenino , Humanos , Hidrogeles/química , Hidrogeles/uso terapéutico , Liposomas , Ratones , Ratones Desnudos , Nanopartículas/química , Nanopartículas/uso terapéutico , Poliésteres/química , Poliésteres/uso terapéutico , Sarcoma/tratamiento farmacológico
4.
Front Immunol ; 12: 706133, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34381456

RESUMEN

The axis of Programmed cell death-1 receptor (PD-1) with its ligand (PD-L1) plays a critical role in colorectal cancer (CRC) in escaping immune surveillance, and blocking this axis has been found to be effective in a subset of patients. Although blocking PD-L1 has been shown to be effective in 5-10% of patients, the majority of the cohorts show resistance to this checkpoint blockade (CB) therapy. Multiple factors assist in the growth of resistance to CB, among which T cell exhaustion and immunosuppressive effects of immune cells in the tumor microenvironment (TME) play a critical role along with other tumor intrinsic factors. We have previously shown the polyketide antibiotic, Mithramycin-A (Mit-A), an effective agent in killing cancer stem cells (CSCs) in vitro and in vivo in a subcutaneous murine model. Since TME plays a pivotal role in CB therapy, we tested the immunomodulatory efficacy of Mit-A with anti-PD-L1 mAb (αPD-L1) combination therapy in an immunocompetent MC38 syngeneic orthotopic CRC mouse model. Tumors and spleens were analyzed by flow cytometry for the distinct immune cell populations affected by the treatment, in addition to RT-PCR for tumor samples. We demonstrated the combination treatment decreases tumor growth, thus increasing the effectiveness of the CB. Mit-A in the presence of αPD-L1 significantly increased CD8+ T cell infiltration and decreased immunosuppressive granulocytic myeloid-derived suppressor cells and anti-inflammatory macrophages in the TME. Our results revealed Mit-A in combination with αPD-L1 has the potential for augmented CB therapy by turning an immunologically "cold" into "hot" TME in CRC.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Inhibidores de Puntos de Control Inmunológico/farmacología , Plicamicina/análogos & derivados , Animales , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos C57BL , Plicamicina/farmacología
5.
Cancer Sci ; 112(9): 3846-3855, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34286904

RESUMEN

Heterogeneous nuclear ribonucleoprotein L-like (HNRNPLL), a suppressor of colorectal cancer (CRC) metastasis, is transcriptionally downregulated when CRC cells undergo epithelial-mesenchymal transition (EMT). Here we show that decrease of MYB mediates the downregulation of HNRNPLL during EMT. The promoter activity was attributed to a region from -273 to -10 base pairs upstream of the transcription start site identified by 5'-RACE analysis, and the region contained potential binding sites for MYB and SP1. Luciferase reporter gene assays and knockdown or knockout experiments for genes encoding the MYB family proteins, MYB, MYBL1, and MYBL2, revealed that MYB was responsible for approximately half of the promoter activity. On the other hand, treatment with mithramycin A, an inhibitor for SP1 and SP3, suppressed the promoter activity and their additive contribution was confirmed by knockout experiments. The expression level of MYB was reduced on EMT while that of SP1 and SP3 was unchanged, suggesting that the downregulation of HNRNPLL during EMT was mediated by the decrease of MYB expression while SP1 and SP3 determine the basal transcription level of HNRNPLL. Histopathological analysis confirmed the accumulation of MYB-downregulated cancer cells at the invasion front of clinical CRC tissues. These results provide an insight into the molecular mechanism underlying CRC progression.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Transición Epitelial-Mesenquimal/genética , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Proteínas Proto-Oncogénicas c-myb/metabolismo , Sitios de Unión , Proliferación Celular/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Progresión de la Enfermedad , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Técnicas de Inactivación de Genes , Células HT29 , Humanos , Metástasis de la Neoplasia , Plicamicina/análogos & derivados , Plicamicina/farmacología , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-myb/genética , Factor de Transcripción Sp1/antagonistas & inhibidores , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo , Transcripción Genética/genética , Transfección
6.
In Vitro Cell Dev Biol Anim ; 57(3): 315-323, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33580416

RESUMEN

Specific protein 1 (SP1) might act as a critical transcription regulator in myocardial infarction (MI), but little evidence about its function in regulating cardiac apoptosis, a major cause of MI development, has been revealed. This study tried to investigate the role of SP1 in MI and its interaction with poly-ADP-ribose polymerase (PARP)-1 by using SP1 inhibitor, mithramycin A (mithA). Primary mouse cardiomyocytes and commercial mouse cardiomyocytes were subjected to mithA treatment under hypoxia conditions, while cell viability, Nix promoter activity, and its expression were detected correspondingly. PARP overexpression and knockdown were conducted, respectively, in mithA-treated and SP1-overexpressing cells. Co-immunoprecipitation was used to verify the interaction between PARP and SP1. For in vivo experiments, mithA administration was performed after the injections of adenovirus for PARP overexpression, and then, MI introduction was carried out. Infarct size and lactate dehydrogenase level were measured to assess MI injury. SP1 inhibitor mithA attenuated hypoxia-induced decrease of cell viability and Nix transcriptional activation, which could be inhibited by PARP overexpression. Knockdown of PARP prevented SP1-induced transcription of Nix and cell viability change, and PARP showed direct interaction with SP1. Furthermore, mithA administration reduced MI injuries, while PARP overexpression could suppress the improvement. The cardioprotective role of SP1 inhibitor mithA was demonstrated here expanding the role of SP1 in MI development involving hypoxia-induced cardiac apoptosis. Moreover, PARP acted as a transcriptional coactivator in Nix transcription involving its interaction with SP1.


Asunto(s)
Cardiotónicos/farmacología , Infarto del Miocardio/patología , Miocitos Cardíacos/patología , Plicamicina/análogos & derivados , Poli(ADP-Ribosa) Polimerasas/metabolismo , Factor de Transcripción Sp1/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Hipoxia de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Plicamicina/farmacología , Unión Proteica/efectos de los fármacos , Factor de Transcripción Sp1/metabolismo , Transcripción Genética/efectos de los fármacos
7.
Oncol Rep ; 45(1): 95-106, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33416166

RESUMEN

The oncogenic role of Erb­B2 Receptor Tyrosine Kinase 2 (ERBB2) has been identified in several types of cancer, but less is known on its function and mechanism of action in cervical cancer cells. The present study employed a multipronged approach to investigate the role of ERBB2 in cervical cancer. ERBB2 and microRNA (miR)­3184­5p expression was assessed in patient­derived cervical cancer biopsy tissues, revealing that higher levels of ERBB2 and lower levels of miR­3184­5p were associated with clinicopathological indicators of cervical cancer progression. Furthermore, ERBB2 stimulated proliferation, migration and sphere­formation of cervical cancer cells in vitro. This effect was mediated by enhanced phosphatidylinositol­4,5­bisphosphate 3­kinase catalytic subunit α activity. Additionally, it was revealed that miR­3184­5p directly suppressed ERBB2 in cervical cancer cells. The p53 activator Mithramycin A stimulated p53 and miR­3184­5p expression, thereby lowering the levels of ERBB2 and attenuating proliferation, migration and sphere­formation of cervical cancer cells. In conclusion, the findings of the present study suggested ERBB2 as an oncogenic protein that may promote invasiveness in cervical cancer cells. Treatment of cervical cancer cells with the p53 activator Mithramycin A restored the levels of the endogenous ERBB2 inhibitor miR­3184­5p and may represent a novel treatment strategy for cervical cancer.


Asunto(s)
MicroARNs/metabolismo , Receptor ErbB-2/genética , Proteína p53 Supresora de Tumor/metabolismo , Neoplasias del Cuello Uterino/genética , Adulto , Anciano , Biopsia , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Cuello del Útero/patología , Cuello del Útero/cirugía , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Histerectomía , MicroARNs/genética , Persona de Mediana Edad , Invasividad Neoplásica , Plicamicina/análogos & derivados , Plicamicina/farmacología , Proteína p53 Supresora de Tumor/agonistas , Neoplasias del Cuello Uterino/patología , Neoplasias del Cuello Uterino/cirugía
8.
Int J Radiat Oncol Biol Phys ; 109(5): 1454-1471, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33373655

RESUMEN

PURPOSE: The oncogenic EWS:Fli1 fusion protein is a key transcriptional mediator of Ewing sarcoma initiation, progression, and therapeutic resistance. Mithramycin A (MithA) is a potent and specific inhibitor of transcription mediated by the EWS:Fli1. We tested the hypothesis that pretreatment with MithA could selectively radiosensitize EWS:Fli1+ tumor cells by altering the transcriptional response to radiation injury. METHODS AND MATERIALS: A panel of 4 EWS:Fli1+ and 3 EWS:Fli1- Ewing sarcoma cell lines and 1 nontumor cell line were subjected to MithA dose-response viability assays to determine the relative potency of MithA in cells possessing or lacking the EWS:Fli1 fusion. Radiosensitization by MithA was evaluated by clonogenic survival assays in vitro and in a murine xenograft model. DNA damage was evaluated by comet assay and γ-H2Ax flow cytometry. Immunoblotting, flow cytometry, and reverse-transcription, polymerase chain reaction were used to evaluate DNA damage-induced signaling and repair processes and apoptosis. RESULTS: We found that MithA alone could potently and selectively inhibit the growth of EWS:Fli1+ tumor cells, but not cells lacking this fusion. Pretreatment with MithA for 24 hours before irradiation significantly reduced clonogenic survival in vitro and delayed tumor regrowth in vivo, prolonging survival of EWS:Fli1+ tumor-bearing mice. Although MithA did not increase the level of DNA double-strand breaks, mechanistic studies revealed that MithA pretreatment selectively inhibited DNA double-strand break repair through downregulation of EWS:Fli1-mediated transcription, leading to tumor cell death by apoptosis. CONCLUSIONS: Our data indicate that MithA is an effective radiosensitizer of EWS:Fli1+ tumors and may achieve better local control at lower doses of radiation.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN/efectos de los fármacos , Proteínas de Fusión Oncogénica/metabolismo , Plicamicina/análogos & derivados , Proteína Proto-Oncogénica c-fli-1/metabolismo , Proteína EWS de Unión a ARN/metabolismo , Fármacos Sensibilizantes a Radiaciones/farmacología , Sarcoma de Ewing/radioterapia , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensayo Cometa , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Histonas/metabolismo , Ratones , Plicamicina/farmacología , Tolerancia a Radiación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sarcoma de Ewing/química , Sarcoma de Ewing/genética , Sarcoma de Ewing/patología , Ensayos Antitumor por Modelo de Xenoinjerto
9.
J Am Heart Assoc ; 9(1): e014103, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31880978

RESUMEN

Background Activation of the YAP (Yes-associated protein) pathway has been demonstrated to be related to smooth muscle cells (SMCs) phenotypic modulation and vessel restenosis. The aim of this study was to illustrate the molecular mechanisms that regulate the expression of YAP during the process of SMCs phenotypic switch. Whether the molecular basis identified in the study could be a potential therapeutic target for drug-eluting stents is further tested. Methods and Results In cell culture and in rat carotid arterial injury models, Sp-1 (specificity protein 1) expression was significantly induced, and correlated with SMCs proliferative phenotype. Overexpression of Sp-1 promoted SMCs proliferation and migration. Conversely, siSp-1 transfection or Sp-1 inhibitor Mithramycin A treatment attenuates SMC proliferation and migration. Through gain- and loss-function assays, we demonstrated that YAP was involved in Sp-1-mediated SMC phenotypic switch. Mechanistically, activated Sp-1 regulated YAP transcriptional expression through binding to its promoter. Moreover, we fabricated a Sp-1 inhibitor Mithramycin A-eluting stent and further tested it. In the rabbit carotid model, Mithramycin A-eluting stent inhibited YAP transcription and attenuated in-stent restenosis through regulating YAP-mediated SMC phenotypic switch. Conclusions Sp-1 controls phenotypic modulation of SMC by regulating transcription factor YAP. Drug-eluting stent targeting Sp-1 might represent a novel therapeutic strategy to prevent in-stent restenosis.


Asunto(s)
Angioplastia de Balón/instrumentación , Proteínas Reguladoras de la Apoptosis/metabolismo , Fármacos Cardiovasculares/farmacología , Traumatismos de las Arterias Carótidas/terapia , Estenosis Carotídea/prevención & control , Plasticidad de la Célula/efectos de los fármacos , Stents Liberadores de Fármacos , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Plicamicina/análogos & derivados , Factor de Transcripción Sp1/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/metabolismo , Arterias Carótidas/patología , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/patología , Estenosis Carotídea/genética , Estenosis Carotídea/metabolismo , Estenosis Carotídea/patología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Masculino , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Fenotipo , Plicamicina/farmacología , Diseño de Prótesis , Conejos , Ratas , Transducción de Señal , Factor de Transcripción Sp1/genética , Proteínas Señalizadoras YAP
10.
Genes (Basel) ; 10(12)2019 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-31783675

RESUMEN

Prenyl diphosphate synthase subunit 2 (PDSS2) is the first key enzyme in the CoQ10 biosynthesis pathway, and contributes to various metabolic and nephritic diseases. It has been reported that PDSS2 is downregulated in several types of tumors and acts as a potential tumor suppressor gene to inhibit the proliferation and migration of cancer cells. However, the regulatory mechanism of PDSS2 expression remains elusive. In the present study, we first identified and characterized the PDSS2 promoter region. We established four different luciferase reporter constructs which mainly cover the 2 kb region upstream of the PDSS2 gene transcription initiation site. Series luciferase reporter assay demonstrated that all four constructs have prominent promoter activity, and the core promoter of PDSS2 is mainly located within the 202 bp region near its transcription initiation site. Transcription factor binding site analysis revealed that the PDSS2 promoter contains binding sites for canonical transcription factors such as Sp1 and GATA-1. Overexpression of Sp1 significantly inhibited PDSS2 promoter activity, as well as its endogenous expression, at both mRNA and protein levels in lung cancer cells. Site-directed mutagenesis assay further confirmed that the Sp1 binding sites are essential for proximal prompter activity of PDSS2. Consistently, a selective Sp1 inhibitor, mithramycin A, treatment repressed the PDSS2 promoter activity, as well as its endogenous expression. Chromatin immunoprecipitation (ChIP) assay revealed that Sp1 binds to the PDSS2 promoter in vivo. Of note, the expression of Sp1 and PDSS2 are negatively correlated, and higher Sp1 expression with low PDSS2 expression is significantly associated with poor prognosis in lung cancer. Taken together, our results strongly suggest the essential role of Sp1 in maintaining the basic constitutive expression of PDSS2, and the pathogenic implication of Sp1-mediated PDSS2 transcriptional repression in lung cancer cells.


Asunto(s)
Transferasas Alquil y Aril/genética , Transferasas Alquil y Aril/metabolismo , Neoplasias Pulmonares/mortalidad , Factor de Transcripción Sp1/genética , Células A549 , Transferasas Alquil y Aril/química , Transferasas Alquil y Aril/efectos de los fármacos , Sitios de Unión , Línea Celular Tumoral , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Plicamicina/análogos & derivados , Plicamicina/farmacología , Pronóstico , Regiones Promotoras Genéticas/efectos de los fármacos , Factor de Transcripción Sp1/metabolismo , Análisis de Supervivencia , Sitio de Iniciación de la Transcripción
11.
Front Immunol ; 10: 2455, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31681329

RESUMEN

B-cell receptor (BCR)-dependent signaling is central for leukemia B-cell homeostasis, as underscored by the promising clinical results obtained in patients with chronic lymphocytic leukemia (CLL) treated with novel agents targeting components of this pathway. Herein, we demonstrate that the mithralog EC-7072 displays high ex vivo cytotoxic activity against leukemia cells from CLL patients independently from high-risk prognostic markers and IGHV mutational status. EC-7072 was significantly less toxic against T cells and NK cells and did not alter the production of the immune effector molecules IFN-γ and perforin. EC-7072 directly triggered caspase-3-dependent CLL cell apoptosis, which was not abrogated by microenvironment-derived factors that sustain leukemia cell survival. RNA-sequencing analyses revealed a dramatic EC-7072-driven reprograming of the transcriptome of CLL cells, including a wide downregulation of multiple components and targets of the BCR signaling pathway. Accordingly, we found decreased levels of phosphorylated signaling nodes downstream of the BCR. Crosslinking-mediated BCR activation antagonized CLL cell death triggered by EC-7072, increased the phosphorylation levels of the abovementioned signaling nodes and upregulated BCL2 expression, suggesting that the mithralog disrupts CLL cell viability by targeting the BCR signaling axis at multiple levels. EC-7072 exerted similar or higher antileukemic activity than that of several available CLL therapies and displayed additive or synergistic interaction with these drugs in killing CLL cells. Overall, our findings provide rationale for future investigation to test whether EC-7072 may be a potential therapeutic option for patients with CLL and other B-cell malignancies.


Asunto(s)
Apoptosis/efectos de los fármacos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Plicamicina/análogos & derivados , Receptores de Antígenos de Linfocitos B/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Antibióticos Antineoplásicos/farmacología , Caspasa 3/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Perfilación de la Expresión Génica/métodos , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Humanos , Interferón gamma/metabolismo , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/metabolismo , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/patología , Fosforilación/efectos de los fármacos , Plicamicina/farmacología , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/metabolismo , Transducción de Señal/genética , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Células Tumorales Cultivadas , Microambiente Tumoral/efectos de los fármacos
12.
Sci Rep ; 9(1): 15202, 2019 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-31645574

RESUMEN

The pivotal role of cancer initiating stem cells (CSCs) in tumor initiation, growth, metastasis and drug resistance has led to the postulation of a 'total cancer therapy' paradigm, which involves targeting both cancer cells and CSCs for effective therapy. However, the progress in identifying drugs for total cancer therapy has been limited. Herein, we show for the first time that mithramycin A (Mit-A) can successfully inhibit CSC proliferation, in addition to inhibiting bulk cancer cells in a model of colorectal cancer (CRC), the second leading cause of death among men and women in the United States. To this end, a polymeric nanofiber scaffold culture system was established to develop 3D tumor organoids (tumoroids) from CRC cell lines such as HT29, HCT116, KM12, CT26 and MC38 as well as ex vivo mouse tumors. These tumoroids possessed increased expression of CSC markers and transcription factors, expanded the number of CSCs in culture and increased CSC functional properties measured by aldehyde dehydrogenase activity. Screening of an NCI library of FDA approved drugs led to the identification of Mit-A as a potential total cancer therapy drug. In both sphere and tumoroid culture, Mit-A inhibits cancer growth by reducing the expression of cancer stemness markers. In addition, Mit-A inhibits the expression of SP1, a previously known target in CRCs. Moreover, Mit-A significantly reduces growth of tumoroids in ex vivo cultures and CRC tumor growth in vivo. Finally, a dose-dependent treatment on CRC cells indicate that Mit-A significantly induces the cell death and PARP-cleavage of both CSC and non-CSC cells. Taken together the results of these in vitro, ex vivo and in vivo studies lead to the inference that Mit-A is a promising drug candidate for total cancer therapy of CRCs.


Asunto(s)
Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Plicamicina/análogos & derivados , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Neoplasias Colorrectales/patología , Células HCT116 , Células HT29 , Humanos , Ratones Endogámicos C57BL , Células Madre Neoplásicas/patología , Plicamicina/farmacología , Plicamicina/uso terapéutico
13.
Oncol Rep ; 42(5): 2016-2028, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31436300

RESUMEN

The restricted expression of Wilms tumor 1 (WT1) and cyclin A1 (CCNA1) in normal tissues, as opposed to their abnormal expression in leukemia demonstrates the applicability of WT1 and CCNA1 as cancer antigens for immunotherapy, and as markers for prognosis and relapse. In this study, the WT1 and CCNA1 mRNA levels were found to be elevated in bone marrow samples from pediatric acute promyelocytic leukemia (APL or AML­M3) patients, and to be quite varied in pediatric acute lymphocytic leukemia (ALL) patients, compared to non­leukemic bone marrow controls. Consistent with the observed upregulation of both WT1 and CCNA1 in APL, WT1 overexpression elevated the CCNA1 mRNA levels in K562 leukemia cells. Treatment with curcumin decreased the WT1 levels in K562 cells, and also decreased CCNA1 protein expression. The examination of the CCNA1 promoter identified potential canonical WT1 binding sites within the 3­kb region upstream of the transcription start site. Chromatin immunoprecipitation and luciferase reporter assays confirmed WT1 binding and the activation of the CCNA1 promoter. Furthermore, the GC­rich core CCNA1 promoter region provided additional non­canonical WT1 activation sites, as revealed by promoter assays. The importance of the GC­rich core region of the CCNA1 promoter was confirmed by treating the K562 cells with mithramycin A, which blocks the binding of zinc finger transcription factors to GC­rich sequences. Mithramycin A subsequently suppressed both CCNA1 promoter activity and protein expression in the K562 cells. Taken together, the data from the WT1 overexpression, and curcumin and mithramycin A treatment experiments, as well as those from chromatin binding assays, along with inferences from patient RNA analyses, establish a plausible link between WT1 and CCNA1, and support the functional significance of an elevated WT1 expression in leukemia, which may also affect CCNA1 expression.


Asunto(s)
Ciclina A1/genética , Leucemia Promielocítica Aguda/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proteínas WT1/genética , Adolescente , Sitios de Unión , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Niño , Preescolar , Curcumina/farmacología , Ciclina A1/química , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Lactante , Células K562 , Leucemia Promielocítica Aguda/metabolismo , Masculino , Plicamicina/análogos & derivados , Plicamicina/farmacología , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Regiones Promotoras Genéticas/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Proteínas WT1/química , Proteínas WT1/metabolismo
14.
Eur Urol Oncol ; 2(4): 415-424, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31277777

RESUMEN

BACKGROUND: The TMPRSS2-ERG gene fusion is the most frequent genetic rearrangement in prostate cancers and results in broad transcriptional reprogramming and major phenotypic changes. Interaction and cooperation of ERG and SP1 may be instrumental in sustaining the tumorigenic and metastatic phenotype and could represent a potential vulnerability in ERG fusion-positive tumors. OBJECTIVE: To test the activity of EC-8042, a compound able to block SP1, in cellular and mouse models of ERG-positive prostate cancer. DESIGN, SETTING, AND PARTICIPANTS: We evaluated the activity of EC-8042 in cell cultures and ERG/PTEN transgenic/knockout mice that provide reliable models for testing novel therapeutics in this specific disease context. Using a new protocol to generate tumor spheroids from ERG/PTEN mice, we also examined the effects of EC-8042 on tumor-propagating stem-like cancer cells with high self-renewal and tumorigenic capabilities. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: The efficacy of EC-8042 was determined by measuring the proliferative capacity and target gene expression in cell cultures, invasive and metastatic capabilities in chick chorioallantoic membrane assays, and tumor development in mice. Significance was determined using statistical test. RESULTS AND LIMITATIONS: EC-8042 blocked transcription of ERG-regulated genes and reverted the invasive and metastatic phenotype of VCaP cells. EC-8042 blocked the expansion of stem-like tumor cells in tumor spheroids from VCaP cells and mouse-derived tumors. In ERG/PTEN mice, systemic treatment with EC-8042 inhibited ERG-regulated gene transcription, tumor progression, and tumor-propagating stem-like tumor cells. CONCLUSIONS: Our data support clinical testing of EC-8042 for the treatment of ERG-positive prostate cancer in precision medicine approaches. PATIENT SUMMARY: In this study, EC-8042, a novel compound with a favorable pharmacological and toxicological profile, exhibited relevant activity in cell cultures and in vivo in a genetically engineered mouse model that closely recapitulates the features of clinically aggressive ERG-positive prostate cancer. Our data indicate that further evaluation of EC-8042 in clinical trials is warranted.


Asunto(s)
Plicamicina/análogos & derivados , Neoplasias de la Próstata/genética , Factor de Transcripción Sp1/antagonistas & inhibidores , Regulador Transcripcional ERG/genética , Animales , Línea Celular Tumoral , Humanos , Masculino , Ratones Transgénicos , Células Madre Neoplásicas , Fosfohidrolasa PTEN/genética , Plicamicina/farmacología , Plicamicina/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico
15.
Neuroscience ; 412: 207-215, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31220545

RESUMEN

High voltage-activated (HVA) Ca2+ (CaV) channels are oligomeric complexes formed by an ion-conducting main subunit (Cavα1) and at least two auxiliary subunits (Cavß and CaVα2δ). It has been reported that the expression of CaVα2δ1 increases in the dorsal root ganglia (DRGs) of animals with mechanical allodynia, and that the transcription factor Sp1 regulates the expression of the auxiliary subunit. Hence, the main aim of this work was to investigate the role of Sp1 as a molecular determinant of the exacerbated expression of CaVα2δ-1 in the nerve ligation-induced model of mechanical allodynia. Our results show that ligation of L5/L6 spinal nerves (SNL) produced allodynia and increased the expression of Sp1 and CaVα2δ-1 in the DRGs. Interestingly, intrathecal administration of the Sp1 inhibitor mithramycin A (Mth) prevented allodynia and decreased the expression of Sp1 and CaVα2δ-1. Likewise, electrophysiological recordings showed that incubation with Mth decreased Ca2+ current density in the DRG neurons, acting mostly on HVA channels. These results suggest that L5/L6 SNL produces mechanical allodynia and increases the expression of the transcription factor Sp1 and the subunit CaVα2δ-1 in the DRGs, while Mth decreases mechanical allodynia and Ca2+ currents through HVA channels in sensory neurons by reducing the functional expression of the CaVα2δ-1 subunit.


Asunto(s)
Canales de Calcio/metabolismo , Ganglios Espinales/metabolismo , Neuralgia/metabolismo , Células Receptoras Sensoriales/metabolismo , Factor de Transcripción Sp1/metabolismo , Animales , Femenino , Ganglios Espinales/efectos de los fármacos , Neuralgia/etiología , Traumatismos de los Nervios Periféricos/complicaciones , Traumatismos de los Nervios Periféricos/metabolismo , Plicamicina/análogos & derivados , Plicamicina/farmacología , Ratas Wistar , Células Receptoras Sensoriales/efectos de los fármacos , Factor de Transcripción Sp1/antagonistas & inhibidores
16.
Biomed Chromatogr ; 33(8): e4544, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30927450

RESUMEN

Mithramycin (MTM) has potent anticancer activity, but severe toxicities restrict its clinical use. Semi-synthetic approaches have yielded novel MTM analogs with potentially lower toxicity and similar efficacy. In an effort to transition these analogs into in vivo models, a bioanalytical method was developed for their quantification in mouse plasma. Here we present the validation of the method for the quantitation of mithramycin SA-tryptophan (MTMSA-Trp) as well as the applicability of the methodology for assaying additional analogs, including MTM, mithramycin SK (MTMSK) and mithramycin SA-phenylalanine (MTMSA-Phe) with run times of 6 min. Assay linearity ranged from 5 to 100 ng/mL. Accuracies of calibration standards and quality control samples were within 15% of nominal with precision variability of <20%. MTMSA-Trp was stable for 30 days at -80°C and for at least three freeze-thaw cycles. Methanol (-80°C) extraction afforded 92% of MTMSA-Trp from plasma. Calibration curves for MTM and analogs were also linear from ≤5 to 100 ng/mL. This versatile method was used to quantitate MTM analogs in plasma samples collected during preclinical pharmacokinetic studies.


Asunto(s)
Cromatografía Líquida de Alta Presión/métodos , Espectrometría de Masas/métodos , Plicamicina/análogos & derivados , Plicamicina/sangre , Animales , Antibióticos Antineoplásicos , Estabilidad de Medicamentos , Femenino , Límite de Detección , Modelos Lineales , Ratones , Plicamicina/química , Plicamicina/farmacocinética , Reproducibilidad de los Resultados
17.
J Hypertens ; 37(5): 964-971, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30335670

RESUMEN

OBJECTIVE: Melatonin, an endogenous neurohormone secreted predominately by the pineal gland, has a variety of physiological functions. However, its protective role in atherosclerosis is not clear. In this study, we sought to investigate the potential effects of melatonin in modulating atherosclerotic plaque stability in apolipoprotein E knockout (ApoE) mice. METHOD AND RESULTS: Smooth muscle cells were treated with melatonin, which significantly increased mRNA and protein levels of a key intracellular enzyme essential for collagen maturation and secretion, prolyl-4-hydroxylase α1 (P4Hα1). Mechanistically, melatonin increased Akt phosphorylation and transcriptional activation of specificity protein 1 (Sp1), which bound with the P4Hα1 promoter and then induced P4Hα1 expression. Pretreatment with either Akt inhibitor LY294002 or Sp1 inhibitor mithramycin A (MTM) could inhibit melatonin-induced P4Hα1 expression. Finally, atherosclerotic lesions were induced by placing a perivascular collar on the right common carotid artery of ApoE mice, which were received with or without different doses of melatonin or MTM. High-dose melatonin enhanced atherosclerotic plaque stability in ApoE mice in vivo by inducing the expression of P4Hα1, which was reversed by MTM. CONCLUSION: We propose that melatonin supplementation may provide a novel and promising approach to atherosclerosis treatment.


Asunto(s)
Apolipoproteínas E/genética , Depresores del Sistema Nervioso Central/farmacología , Expresión Génica/efectos de los fármacos , Melatonina/farmacología , Miocitos del Músculo Liso/metabolismo , Procolágeno-Prolina Dioxigenasa/genética , Animales , Línea Celular , Depresores del Sistema Nervioso Central/administración & dosificación , Cromonas/farmacología , Inhibidores Enzimáticos/farmacología , Masculino , Melatonina/administración & dosificación , Ratones , Ratones Noqueados , Morfolinas/farmacología , Fosforilación/efectos de los fármacos , Placa Aterosclerótica/tratamiento farmacológico , Plicamicina/análogos & derivados , Plicamicina/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/metabolismo , Factor de Transcripción Sp1/antagonistas & inhibidores , Factor de Transcripción Sp1/genética
18.
J Gen Physiol ; 150(12): 1595, 2018 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-30455181

RESUMEN

JGP study explains how chromomycin A2 affects insulin secretion.


Asunto(s)
Células Secretoras de Insulina , Islotes Pancreáticos , Glucosa , Insulina , Secreción de Insulina , Fosfolipasas A2 , Plicamicina/análogos & derivados
19.
Neurochem Int ; 121: 59-68, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30273642

RESUMEN

Numerous studies have focused on the deteriorate role of amyloid-ß (Aß) on retina, implying the potential pathogenic mechanism underlying age-related macular degeneration (AMD). However, the mechanism underlying the Aß deposition in AMD patients remains unknown. Beta-site amyloid precursor protein-cleaving enzyme 1 (BACE1), rate-limiting enzyme for Aß production, plays an important role in Aß deposition in the brain. In the current study, we aimed to clarify the regulation mechanism of BACE1 and explore potential drug targets using a lipofuscinfluorophore A2E-mediated photo-oxidation model. In this model, Aß1-40 and Aß1-42 levels increased simultaneously with the enhanced BACE1 expression. These changes were associated with the hypomethylation of specific loci within the BACE1 gene promoter and the decreased levels of DNA methyltransferase 1 (DNMT1). Furthermore, we noticed overlapping regions of differentially methylated CpG islands and specificity protein (Sp1) binding sites within the BACE1 promoter. We employed chromatin immunoprecipitation (ChIP) assay to verify that the decreased BACE1 promoter methylation by DNMT1 enabled increased binding between Sp1 and the BACE1 promoter, which further enhanced BACE1 transcription. The inhibition of Sp1 with mithramycin A (MTM) could down-regulate the expression of BACE1 as well as alleviate the RPE barrier morphology and function impairment. Our results for the first time show the competitive regulation of BACE1 by transcription factor Sp1 and DNMT1 after photo-oxidation and confirm the potential novel protective role of MTM on RPE cells.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/biosíntesis , Ácido Aspártico Endopeptidasas/biosíntesis , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Estimulación Luminosa/efectos adversos , Epitelio Pigmentado de la Retina/metabolismo , Retinoides/toxicidad , Factor de Transcripción Sp1/metabolismo , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/genética , Péptidos beta-Amiloides/metabolismo , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/genética , Secuencia de Bases , Línea Celular , Relación Dosis-Respuesta a Droga , Expresión Génica , Humanos , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Plicamicina/análogos & derivados , Plicamicina/farmacología , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/patología , Pigmentos Retinianos/toxicidad
20.
J Gen Physiol ; 150(12): 1747-1757, 2018 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-30352794

RESUMEN

Modulators of insulin secretion could be used to treat diabetes and as tools to investigate ß cell regulatory pathways in order to increase our understanding of pancreatic islet function. Toward this goal, we previously used an insulin-linked luciferase that is cosecreted with insulin in MIN6 ß cells to perform a high-throughput screen of natural products for chronic effects on glucose-stimulated insulin secretion. In this study, using multiple phenotypic analyses, we found that one of the top natural product hits, chromomycin A2 (CMA2), potently inhibited insulin secretion by at least three potential mechanisms: disruption of Wnt signaling, interference of ß cell gene expression, and partial suppression of Ca2+ influx. Chronic treatment with CMA2 largely ablated glucose-stimulated insulin secretion even after washout, but it did not inhibit glucose-stimulated generation of ATP or Ca2+ influx. However, by using the KATP channel opener diazoxide, we uncovered defects in depolarization-induced Ca2+ influx that may contribute to the suppressed secretory response. Glucose-responsive ERK1/2 and S6 phosphorylation were also disrupted by chronic CMA2 treatment. By querying the FUSION bioinformatic database, we revealed that the phenotypic effects of CMA2 cluster with a number of Wnt-GSK3 pathway-related genes. Furthermore, CMA2 consistently decreased GSK3ß phosphorylation and suppressed activation of a ß-catenin activity reporter. CMA2 and a related compound, mithramycin, are known to have DNA interaction properties, possibly abrogating transcription factor binding to critical ß cell gene promoters. We observed that CMA2 but not mithramycin suppressed expression of PDX1 and UCN3. However, neither expression of INSI/II nor insulin content was affected by chronic CMA2. The mechanisms of CMA2-induced insulin secretion defects may involve components both proximal and distal to Ca2+ influx. Therefore, CMA2 is an example of a chemical that can simultaneously disrupt ß cell function through both noncytotoxic and cytotoxic mechanisms. Future therapeutic applications of CMA2 and similar aureolic acid analogues should consider their potential effects on pancreatic islet function.


Asunto(s)
Secreción de Insulina/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Plicamicina/análogos & derivados , Animales , Línea Celular , Expresión Génica/efectos de los fármacos , Humanos , Ratones , Plicamicina/aislamiento & purificación , Plicamicina/farmacología , Cultivo Primario de Células , Transducción de Señal/efectos de los fármacos , Streptomyces/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...