Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Methods Mol Biol ; 2612: 157-168, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36795366

RESUMEN

Multiplex arrays designed for enzyme-linked immunosorbent assays (ELISAs) are robust and cost-effective for profiling biomarkers. Identification of relevant biomarkers in biological matrices or fluids helps in the understanding of disease pathogenesis. Here, we describe a sandwich ELISA-based multiplex assay to assess growth factor and cytokine levels in cerebrospinal fluid (CSF) samples derived from multiple sclerosis patients, amyotrophic lateral sclerosis patients, and control subjects without any neurological disorder. Results indicate that multiplex assay designed for the sandwich ELISA method is a unique, robust, and cost-effective method for profiling growth factors and cytokines present in CSF samples.


Asunto(s)
Esclerosis Amiotrófica Lateral , Citocinas , Humanos , Citocinas/líquido cefalorraquídeo , Péptidos y Proteínas de Señalización Intercelular , Ensayo de Inmunoadsorción Enzimática/métodos , Biomarcadores/líquido cefalorraquídeo
2.
Biomolecules ; 12(4)2022 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-35454092

RESUMEN

The plasminogen activation system regulates the activity of the serine protease, plasmin. The role of plasminogen receptors in cancer progression is being increasingly appreciated as key players in modulation of the tumor microenvironment. The interaction of plasminogen with cells to promote plasminogen activation requires the presence of proteins exposing C-terminal lysines on the cell surface. Plg-RKT is a structurally unique plasminogen receptor because it is an integral membrane protein that is synthesized with and binds plasminogen via a C-terminal lysine exposed on the cell surface. Here, we have investigated the expression of Plg-RKT in human breast tumors and human breast cancer cell lines. Breast cancer progression tissue microarrays were probed with anti-Plg-RKT mAB and we found that Plg-RKT is widely expressed in human breast tumors, that its expression is increased in tumors that have spread to draining lymph nodes and distant organs, and that Plg-RKT expression is most pronounced in hormone receptor (HR)-positive tumors. Plg-RKT was detected by Western blotting in human breast cancer cell lines. By flow cytometry, Plg-RKT cell surface expression was highest on the most aggressive tumor cell line. Future studies are warranted to address the functions of Plg-RKT in breast cancer.


Asunto(s)
Neoplasias de la Mama , Receptores de Superficie Celular , Neoplasias de la Mama/genética , Membrana Celular/metabolismo , Femenino , Humanos , Plasminógeno/metabolismo , Receptores de Superficie Celular/genética , Serina Proteasas , Microambiente Tumoral
3.
Nat Commun ; 7: 11980, 2016 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-27351915

RESUMEN

Traumatic brain injury (TBI) is a major health and socio-economic problem, but no pharmacological agent is currently approved for the treatment of acute TBI. Thus, there is a great need for advances in this field. Here, we describe a short peptide (sequence CAQK) identified by in vivo phage display screening in mice with acute brain injury. The CAQK peptide selectively binds to injured mouse and human brain, and systemically injected CAQK specifically homes to sites of brain injury in mouse models. The CAQK target is a proteoglycan complex upregulated in brain injuries. Coupling to CAQK increased injury site accumulation of systemically administered molecules ranging from a drug-sized molecule to nanoparticles. CAQK-coated nanoparticles containing silencing oligonucleotides provided the first evidence of gene silencing in injured brain parenchyma by systemically administered siRNA. These findings present an effective targeting strategy for the delivery of therapeutics in clinical management of acute brain injuries.


Asunto(s)
Lesiones Traumáticas del Encéfalo/terapia , Sistemas de Liberación de Medicamentos , Péptidos , Anciano , Animales , Lesiones Traumáticas del Encéfalo/diagnóstico por imagen , Matriz Extracelular/metabolismo , Humanos , Masculino , Ratones , Persona de Mediana Edad
4.
Methods Mol Biol ; 1314: 287-301, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26139277

RESUMEN

A variation of immunoblotting method (the "Rainbow Western"), permits sequential detection of multiple antigens (MAD) on a single protein blot without stripping off prior antibodies. Because no stripping is involved, immobilized proteins are not lost from the membrane, thus allowing for multiple reprobings of the same membrane with different primary antibodies (≥12), retaining strong signal intensities for all sequential antibody probings. The procedure utilizes horseradish peroxidase (HRPase)-based detection with both a chemiluminescent and colorimetric substrate. Initial incubation of the blot with secondary antibody followed by colorimetric development prior to probing the blot with primary antibodies markedly reduces background in ECL-based detection procedures and permits sequential use of antibodies derived from a single species. In the "Rainbow Western," four different HRPase-colorimetric substrates that produce black, brown, red, and green colors are employed sequentially for detection and simultaneous display of four different antigens on the same membrane. By allowing large amounts of data to be obtained from a single blot, the MAD-immunoblotting and Rainbow Western methods have the potential for researchers to compare the expression of several proteins within a single biological sample. Both techniques could be particularly valuable for analysis of cellular populations that are difficult to isolate in large numbers or of clinical specimens where the amounts of protein samples is minute or only available on a one-time basis.


Asunto(s)
Antígenos/análisis , Western Blotting/métodos , Animales , Anticuerpos/química , Colorimetría/métodos , Peroxidasa de Rábano Silvestre/química , Humanos , Ratones , Conejos
5.
Methods Mol Biol ; 1254: 181-96, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25431066

RESUMEN

Light microscopy allows for the inexpensive and fast detection of neuronal /glial cell demise and estimation of infarct and traumatic lesion volumes; the direct correlates of cell death. Quantitative assessment of brain tissue damage following stroke, traumatic brain injury (TBI ) or neurodegenerative diseases, and recovery after therapeutic intervention has been facilitated by recent developments in computer-assisted image analysis technologies that enable more objective and accurate morphometric quantification of cell injury in whole brain sections. In this chapter, the proposed workflow describes what tasks need to be fulfilled to visualize and gauge cell death characterization by histological stains and immunohistochemical markers.


Asunto(s)
Lesiones Encefálicas/patología , Muerte Celular/genética , Inmunohistoquímica/métodos , Neuronas/patología , Algoritmos , Humanos , Procesamiento de Imagen Asistido por Computador , Biología Molecular/métodos , Neuronas/metabolismo
6.
J Neuroinflammation ; 11: 39, 2014 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-24593993

RESUMEN

BACKGROUND: Traumatic brain injury (TBI) enhances pro-inflammatory responses, neuronal loss and long-term behavioral deficits. Caveolins (Cavs) are regulators of neuronal and glial survival signaling. Previously we showed that astrocyte and microglial activation is increased in Cav-1 knock-out (KO) mice and that Cav-1 and Cav-3 modulate microglial morphology. We hypothesized that Cavs may regulate cytokine production after TBI. METHODS: Controlled cortical impact (CCI) model of TBI (3 m/second; 1.0 mm depth; parietal cortex) was performed on wild-type (WT; C57Bl/6), Cav-1 KO, and Cav-3 KO mice. Histology and immunofluorescence microscopy (lesion volume, glia activation), behavioral tests (open field, balance beam, wire grip, T-maze), electrophysiology, electron paramagnetic resonance, membrane fractionation, and multiplex assays were performed. Data were analyzed by unpaired t tests or analysis of variance (ANOVA) with post-hoc Bonferroni's multiple comparison. RESULTS: CCI increased cortical and hippocampal injury and decreased expression of MLR-localized synaptic proteins (24 hours), enhanced NADPH oxidase (Nox) activity (24 hours and 1 week), enhanced polysynaptic responses (1 week), and caused hippocampal-dependent learning deficits (3 months). CCI increased brain lesion volume in both Cav-3 and Cav-1 KO mice after 24 hours (P < 0.0001, n = 4; one-way ANOVA). Multiplex array revealed a significant increase in expression of IL-1ß, IL-9, IL-10, KC (keratinocyte chemoattractant), and monocyte chemoattractant protein 1 (MCP-1) in ipsilateral hemisphere and IL-9, IL-10, IL-17, and macrophage inflammatory protein 1 alpha (MIP-1α) in contralateral hemisphere of WT mice after 4 hours. CCI increased IL-2, IL-6, KC and MCP-1 in ipsilateral and IL-6, IL-9, IL-17 and KC in contralateral hemispheres in Cav-1 KO and increased all 10 cytokines/chemokines in both hemispheres except for IL-17 (ipsilateral) and MIP-1α (contralateral) in Cav-3 KO (versus WT CCI). Cav-3 KO CCI showed increased IL-1ß, IL-9, KC, MCP-1, MIP-1α, and granulocyte-macrophage colony-stimulating factor in ipsilateral and IL-1ß, IL-2, IL-9, IL-10, and IL-17 in contralateral hemispheres (P = 0.0005, n = 6; two-way ANOVA) compared to Cav-1 KO CCI. CONCLUSION: CCI caused astrocyte and microglial activation and hippocampal neuronal injury. Cav-1 and Cav-3 KO exhibited enhanced lesion volume and cytokine/chemokine production after CCI. These findings suggest that Cav isoforms may regulate neuroinflammatory responses and neuroprotection following TBI.


Asunto(s)
Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/patología , Encéfalo/patología , Caveolina 1/deficiencia , Caveolina 3/deficiencia , Encefalitis/complicaciones , Animales , Caveolina 1/genética , Caveolina 3/genética , Células Cultivadas , Trastornos del Conocimiento/etiología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalitis/genética , Lateralidad Funcional , Hipocampo/citología , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Trastornos del Movimiento/etiología , NADPH Oxidasas/metabolismo , Neuronas/fisiología , Sinaptosomas/metabolismo , Sinaptosomas/patología
7.
FASEB J ; 28(4): 1854-69, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24421398

RESUMEN

Patients with congenital disorder of glycosylation (CDG), type Ib (MPI-CDG or CDG-Ib) have mutations in phosphomannose isomerase (MPI) that impair glycosylation and lead to stunted growth, liver dysfunction, coagulopathy, hypoglycemia, and intestinal abnormalities. Mannose supplements correct hypoglycosylation and most symptoms by providing mannose-6-P (Man-6-P) via hexokinase. We generated viable Mpi hypomorphic mice with residual enzymatic activity comparable to that of patients, but surprisingly, these mice appeared completely normal except for modest (~15%) embryonic lethality. To overcome this lethality, pregnant dams were provided 1-2% mannose in their drinking water. However, mannose further reduced litter size and survival to weaning by 40 and 66%, respectively. Moreover, ~50% of survivors developed eye defects beginning around midgestation. Mannose started at birth also led to eye defects but had no effect when started after eye development was complete. Man-6-P and related metabolites accumulated in the affected adult eye and in developing embryos and placentas. Our results demonstrate that disturbing mannose metabolic flux in mice, especially during embryonic development, induces a highly specific, unanticipated pathological state. It is unknown whether mannose is harmful to human fetuses during gestation; however, mothers who are at risk for having MPI-CDG children and who consume mannose during pregnancy hoping to benefit an affected fetus in utero should be cautious.


Asunto(s)
Ceguera/etiología , Suplementos Dietéticos/toxicidad , Manosa-6-Fosfato Isomerasa/metabolismo , Manosa/toxicidad , Animales , Ceguera/genética , Ceguera/metabolismo , Western Blotting , Células Cultivadas , Trastornos Congénitos de Glicosilación/genética , Trastornos Congénitos de Glicosilación/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Ojo/embriología , Ojo/crecimiento & desarrollo , Ojo/metabolismo , Femenino , Humanos , Inmunohistoquímica , Masculino , Manosa/sangre , Manosa/metabolismo , Manosa-6-Fosfato Isomerasa/genética , Manosafosfatos/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Placenta/efectos de los fármacos , Placenta/embriología , Placenta/metabolismo , Embarazo
8.
Oncotarget ; 4(8): 1212-29, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23902736

RESUMEN

We previously identified SMIP004 (N-(4-butyl-2-methyl-phenyl) acetamide) as a novel inducer of cancer-cell selective apoptosis of human prostate cancer cells. SMIP004 decreased the levels of positive cell cycle regulators, upregulated cyclin-dependent kinase inhibitors, and resulted in G1 arrest, inhibition of colony formation in soft agar, and cell death. However, the mechanism of SMIP004-induced cancer cell selective apoptosis remained unknown. Here, we used chemical genomic and proteomic profiling to unravel a SMIP004-induced pro-apoptotic pathway, which initiates with disruption of mitochondrial respiration leading to oxidative stress. This, in turn, activates two pathways, one eliciting cell cycle arrest by rapidly targeting cyclin D1 for proteasomal degradation and driving the transcriptional downregulation of the androgen receptor, and a second pathway that activates pro-apoptotic signaling through MAPK activation downstream of the unfolded protein response (UPR). SMIP004 potently inhibits the growth of prostate and breast cancer xenografts in mice. Our data suggest that SMIP004, by inducing mitochondrial ROS formation, targets specific sensitivities of prostate cancer cells to redox and bioenergetic imbalances that can be exploited in cancer therapy.


Asunto(s)
Acetamidas/farmacología , Mitocondrias/efectos de los fármacos , Neoplasias de la Próstata/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Ciclina D1/metabolismo , Expresión Génica/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mitocondrias/genética , Mitocondrias/metabolismo , Estrés Oxidativo/efectos de los fármacos , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Distribución Aleatoria , Transducción de Señal/efectos de los fármacos , Ubiquitina/metabolismo , Respuesta de Proteína Desplegada/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Pigment Cell Melanoma Res ; 26(1): 136-42, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23035722

RESUMEN

To date, there are no effective therapies for tumors bearing NRAS mutations, which are present in 15-20% of human melanomas. Here we extend our earlier studies where we demonstrated that the small molecule BI-69A11 inhibits the growth of melanoma cell lines. Gene expression analysis revealed the induction of interferon- and cell death-related genes that were associated with responsiveness of melanoma cell lines to BI-69A11. Strikingly, the administration of BI-69A11 inhibited melanoma development in genetically modified mice bearing an inducible form of activated Nras and a deletion of the Ink4a gene (Nras((Q61K)) ::Ink4a(-/-) ). Biweekly administration of BI-69A11 starting at 10 weeks or as late as 24 weeks after the induction of mutant Nras expression inhibited melanoma development (100 and 36%, respectively). BI-69A11 treatment did not inhibit the development of histiocytic sarcomas, which constitute about 50% of the tumors in this model. BI-69A11-resistant Nras((Q61K)) ::Ink4a(-/-) tumors exhibited increased CD45 expression, reflective of immune cell infiltration and upregulation of gene networks associated with the cytoskeleton, DNA damage response, and small molecule transport. The ability to attenuate the development of NRAS mutant melanomas supports further development of BI-69A11 for clinical assessment.


Asunto(s)
Sustitución de Aminoácidos/genética , Bencimidazoles/uso terapéutico , Inhibidor p16 de la Quinasa Dependiente de Ciclina/deficiencia , Melanoma/tratamiento farmacológico , Lesiones Precancerosas/tratamiento farmacológico , Quinolonas/uso terapéutico , Neoplasias Cutáneas/tratamiento farmacológico , Proteínas ras/genética , Animales , Bencimidazoles/administración & dosificación , Bencimidazoles/farmacología , Proliferación Celular/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inyecciones Intraperitoneales , Antígeno Ki-67/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Melanoma/genética , Melanoma/patología , Ratones , Lesiones Precancerosas/genética , Lesiones Precancerosas/patología , Proteínas Proto-Oncogénicas B-raf/genética , Quinolonas/administración & dosificación , Quinolonas/farmacología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Bibliotecas de Moléculas Pequeñas/farmacología , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Proc Natl Acad Sci U S A ; 108(46): 18637-42, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22049339

RESUMEN

Molecules differentially expressed in blood vessels among organs or between damaged and normal tissues, are attractive therapy targets; however, their identification within the human vasculature is challenging. Here we screened a peptide library in cancer patients to uncover ligand-receptors common or specific to certain vascular beds. Surveying ~2.35 x 10(6) motifs recovered from biopsies yielded a nonrandom distribution, indicating that systemic tissue targeting is feasible. High-throughput analysis by similarity search, protein arrays, and affinity chromatography revealed four native ligand-receptors, three of which were previously unrecognized. Two are shared among multiple tissues (integrin α4/annexin A4 and cathepsin B/apolipoprotein E3) and the other two have a restricted and specific distribution in normal tissue (prohibitin/annexin A2 in white adipose tissue) or cancer (RAGE/leukocyte proteinase-3 in bone metastases). These findings provide vascular molecular markers for biotechnology and medical applications.


Asunto(s)
Vasos Sanguíneos/metabolismo , Médula Ósea/metabolismo , Neoplasias/metabolismo , Secuencias de Aminoácidos , Anexina A4/biosíntesis , Apolipoproteína E3/biosíntesis , Biopsia , Catepsina B/biosíntesis , Regulación Neoplásica de la Expresión Génica , Humanos , Integrina alfa4/biosíntesis , Ligandos , Neovascularización Patológica , Obesidad/metabolismo , Biblioteca de Péptidos
11.
PLoS One ; 6(9): e24341, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21957448

RESUMEN

BACKGROUND: Acute brain injury is an important health problem. Given the critical position of caspase 8 at the crossroads of cell death pathways, we generated a new viable mouse line (Ncasp8(-/-)), in which the gene encoding caspase 8 was selectively deleted in neurons by cre-lox system. METHODOLOGY/PRINCIPAL FINDINGS: Caspase 8 deletion reduced rates of neuronal cell death in primary neuronal cultures and in whole brain organotypic coronal slice cultures prepared from 4 and 8 month old mice and cultivated up to 14 days in vitro. Treatments of cultures with recombinant murine TNFα (100 ng/ml) or TRAIL (250 ng/mL) plus cyclohexamide significantly protected neurons against cell death induced by these apoptosis-inducing ligands. A protective role of caspase 8 deletion in vivo was also demonstrated using a controlled cortical impact (CCI) model of traumatic brain injury (TBI) and seizure-induced brain injury caused by kainic acid (KA). Morphometric analyses were performed using digital imaging in conjunction with image analysis algorithms. By employing virtual images of hundreds of brain sections, we were able to perform quantitative morphometry of histological and immunohistochemical staining data in an unbiased manner. In the TBI model, homozygous deletion of caspase 8 resulted in reduced lesion volumes, improved post-injury motor performance, superior learning and memory retention, decreased apoptosis, diminished proteolytic processing of caspases and caspase substrates, and less neuronal degeneration, compared to wild type, homozygous cre, and caspase 8-floxed control mice. In the KA model, Ncasp8(-/-) mice demonstrated superior survival, reduced seizure severity, less apoptosis, and reduced caspase 3 processing. Uninjured aged knockout mice showed improved learning and memory, implicating a possible role for caspase 8 in cognitive decline with aging. CONCLUSIONS: Neuron-specific deletion of caspase 8 reduces brain damage and improves post-traumatic functional outcomes, suggesting an important role for this caspase in pathophysiology of acute brain trauma.


Asunto(s)
Lesiones Encefálicas/enzimología , Caspasa 8/genética , Corteza Cerebral/efectos de los fármacos , Eliminación de Gen , Ácido Kaínico/toxicidad , Neuronas/efectos de los fármacos , Neuronas/enzimología , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Lesiones Encefálicas/inducido químicamente , Lesiones Encefálicas/patología , Lesiones Encefálicas/fisiopatología , Caspasa 8/metabolismo , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Células Cultivadas , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Corteza Cerebral/fisiopatología , Embrión de Mamíferos , Gliosis/complicaciones , Inflamación/complicaciones , Memoria/efectos de los fármacos , Memoria/fisiología , Ratones , Neuronas/metabolismo , Neuronas/patología , Neurotoxinas/toxicidad , Convulsiones/inducido químicamente , Convulsiones/complicaciones , Factor de Necrosis Tumoral alfa/farmacología
12.
J Biol Chem ; 286(38): 33125-33, 2011 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-21795689

RESUMEN

Neurotransmitter release by catecholaminergic cells is negatively regulated by prohormone cleavage products formed from plasmin-mediated proteolysis. Here, we investigated the expression and subcellular localization of Plg-R(KT), a novel plasminogen receptor, and its role in catecholaminergic cell plasminogen activation and regulation of catecholamine release. Prominent staining with anti-Plg-R(KT) mAb was observed in adrenal medullary chromaffin cells in murine and human tissue. In Western blotting, Plg-R(KT) was highly expressed in bovine adrenomedullary chromaffin cells, human pheochromocytoma tissue, PC12 pheochromocytoma cells, and murine hippocampus. Expression of Plg-R(KT) fused in-frame to GFP resulted in targeting of the GFP signal to the cell membrane. Phase partitioning, co-immunoprecipitation with urokinase-type plasminogen activator receptor (uPAR), and FACS analysis with antibody directed against the C terminus of Plg-R(KT) were consistent with Plg-R(KT) being an integral plasma membrane protein on the surface of catecholaminergic cells. Cells stably overexpressing Plg-R(KT) exhibited substantial enhancement of plasminogen activation, and antibody blockade of non-transfected PC12 cells suppressed plasminogen activation. In functional secretion assays, nicotine-evoked [(3)H]norepinephrine release from cells overexpressing Plg-R(KT) was markedly decreased (by 51 ± 2%, p < 0.001) when compared with control transfected cells, and antibody blockade increased [(3)H]norepinephrine release from non-transfected PC12 cells. In summary, Plg-R(KT) is present on the surface of catecholaminergic cells and functions to stimulate plasminogen activation and modulate catecholamine release. Plg-R(KT) thus represents a new mechanism and novel control point for regulating the interface between plasminogen activation and neurosecretory cell function.


Asunto(s)
Catecolaminas/metabolismo , Plasminógeno/metabolismo , Receptores de Superficie Celular/metabolismo , Médula Suprarrenal/citología , Médula Suprarrenal/efectos de los fármacos , Médula Suprarrenal/metabolismo , Animales , Anticuerpos/farmacología , Bovinos , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Activación Enzimática/efectos de los fármacos , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Ratones , Células PC12 , Transporte de Proteínas/efectos de los fármacos , Ratas , Receptores de Superficie Celular/antagonistas & inhibidores
13.
Pigment Cell Melanoma Res ; 24(4): 703-13, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21592316

RESUMEN

In melanoma, the activation of pro-survival signaling pathways, such as the AKT and NF-κB pathways, is critical for tumor growth. We have recently reported that the AKT inhibitor BI-69A11 causes efficient inhibition of melanoma growth. Here, we show that in addition to its AKT inhibitory activity, BI-69A11 also targets the NF-κB pathway. In melanoma cell lines, BI-69A11 inhibited TNF-α-stimulated IKKα/ß and IκB phosphorylation as well as NF-κB reporter gene expression. Furthermore, the effective inhibition of melanoma growth by BI-69A11 was attenuated upon NF-κB activation. Mechanistically, reduced NF-κB signaling by BI-69-A11 is mediated by the inhibition of sphingosine kinase 1, identified in a screen of 315 kinases. Significantly, we demonstrate that BI-69A11 is well tolerated and orally active against UACC 903 and SW1 melanoma xenografts. Our results demonstrate that BI-69A11 inhibits both the AKT and the NF-κB pathways and that the dual targeting of these pathways may be efficacious as a therapeutic strategy in melanoma.


Asunto(s)
Bencimidazoles/uso terapéutico , Melanoma/tratamiento farmacológico , Terapia Molecular Dirigida , FN-kappa B/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Quinolonas/uso terapéutico , Transducción de Señal/efectos de los fármacos , Neoplasias Cutáneas/tratamiento farmacológico , Administración Oral , Animales , Bencimidazoles/administración & dosificación , Bencimidazoles/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Melanoma/enzimología , Ratones , Ratones Desnudos , FN-kappa B/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Quinolonas/administración & dosificación , Quinolonas/farmacología , Neoplasias Cutáneas/enzimología , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Brain Res ; 1370: 227-37, 2011 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-21075086

RESUMEN

Bax-Inhibitor-1 (BI-1) is an evolutionarily conserved cytoprotective protein that resides in membranes of the endoplasmic reticulum (ER). BI-1's cytoprotective activity is manifested in the context of ER stress, with previous studies showing that BI-1 modulates several ER-associated functions, including Unfolded Protein Response (UPR) signaling. Here we investigated the role of BI-1 in neuroprotection by generating transgenic mice in which BI-1 was constitutively expressed from a neuronal-specific promoter. Cultured primary cortical neurons from BI-1 transgenic mouse embryos exhibited greater resistance to cell death induced by agents known to cause ER stress compared to their non-transgenic counterparts. While brain morphology and vasculature of BI-1 mice appeared to be unchanged from normal non-transgenic mice, BI-1 transgenic mice showed reduced brain lesion volumes and better performance in motoric tests, compared with non-transgenic littermates, in two models of acute brain injury: stroke caused by middle cerebral artery occlusion (MCAO) and traumatic brain injury (TBI) caused by controlled cortical impact. Furthermore, brain tissue from BI-1 transgenic mice showed reduced levels of apoptotic cells and reduced induction of markers of ER stress after brain injury, including CHOP protein expression. In summary, our findings demonstrate that enforced neuronal expression of BI-1 reduces ER stress and provides protection from acute brain injury, suggesting that strategies for enhancing BI-1 expression or activity should be considered for development of new therapies for counteracting the consequences of stroke and acute brain trauma.


Asunto(s)
Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/prevención & control , Retículo Endoplásmico/metabolismo , Proteínas de la Membrana/fisiología , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/prevención & control , Respuesta de Proteína Desplegada/fisiología , Animales , Lesiones Encefálicas/genética , Células Cultivadas , Citoprotección/genética , Citoprotección/fisiología , Retículo Endoplásmico/genética , Humanos , Proteínas de la Membrana/genética , Ratones , Ratones Transgénicos , Transducción de Señal/genética , Accidente Cerebrovascular/genética , Respuesta de Proteína Desplegada/genética
15.
Cancer Cell ; 18(1): 23-38, 2010 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-20609350

RESUMEN

Neuroendocrine (NE) phenotype, seen in >30% of prostate adenocarcinomas (PCa), and NE prostate tumors are implicated in aggressive prostate cancer. Formation of NE prostate tumors in the TRAMP mouse model was suppressed in mice lacking the ubiquitin ligase Siah2, which regulates HIF-1alpha availability. Cooperation between HIF-1alpha and FoxA2, a transcription factor expressed in NE tissue, promotes recruitment of p300 to transactivate select HIF-regulated genes, Hes6, Sox9, and Jmjd1a. These HIF-regulated genes are highly expressed in metastatic PCa and required for hypoxia-mediated NE phenotype, metastasis in PCa, and the formation of NE tumors. Tissue-specific expression of FoxA2 combined with Siah2-dependent HIF-1alpha availability enables a transcriptional program required for NE prostate tumor development and NE phenotype in PCa.


Asunto(s)
Adenocarcinoma/patología , Factor Nuclear 3-beta del Hepatocito/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Tumores Neuroendocrinos/patología , Sistemas Neurosecretores/patología , Neoplasias de la Próstata/patología , Ubiquitina-Proteína Ligasas/fisiología , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Animales , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Factor Nuclear 3-beta del Hepatocito/genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neoplasias Hepáticas Experimentales/genética , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/secundario , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Metástasis Linfática , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/metabolismo , Sistemas Neurosecretores/metabolismo , Fenotipo , Próstata/metabolismo , Próstata/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Transducción de Señal , Activación Transcripcional
16.
J Biol Chem ; 285(29): 22426-36, 2010 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-20421299

RESUMEN

Mixed lineage kinases (MLKs) have been implicated in cytokine signaling as well as in cell death pathways. Our studies show that MLK3 is activated in leukocyte-infiltrated islets of non-obese diabetic mice and that MLK3 activation compromises mitochondrial integrity and induces apoptosis of beta cells. Using an ex vivo model of islet-splenocyte co-culture, we show that MLK3 mediates its effects via the pseudokinase TRB3, a mammalian homolog of Drosophila Tribbles. TRB3 expression strongly coincided with conformational change and mitochondrial translocation of BAX. Mechanistically, MLK3 directly interacted with and stabilized TRB3, resulting in inhibition of Akt, a strong suppressor of BAX translocation and mitochondrial membrane permeabilization. Accordingly, attenuation of MLK3 or TRB3 expression each prevented cytokine-induced BAX conformational change and attenuated the progression to apoptosis. We conclude that MLKs compromise mitochondrial integrity and suppress cellular survival mechanisms via TRB3-dependent inhibition of Akt.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Citocinas/farmacología , Células Secretoras de Insulina/enzimología , Células Secretoras de Insulina/patología , Quinasas Quinasa Quinasa PAM/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas Represoras/metabolismo , Adulto , Animales , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Técnicas de Cocultivo , Activación Enzimática/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Células Secretoras de Insulina/efectos de los fármacos , Ratones , Unión Proteica/efectos de los fármacos , Conformación Proteica , Estabilidad Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteína X Asociada a bcl-2/química , Proteína X Asociada a bcl-2/metabolismo , Proteina Quinasa Quinasa Quinasa 11 Activada por Mitógeno
17.
Cancer Res ; 70(8): 3287-98, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20388784

RESUMEN

Small GTPase Ras homologue enriched in brain (RHEB) binds and activates the key metabolic regulator mTORC1, which has an important role in cancer cells, but the role of RHEB in cancer pathogenesis has not been shown. By performing a meta-analysis of published cancer cytogenetic and transcriptome databases, we defined a gain of chromosome 7q36.1-q36.3 containing the RHEB locus, an overexpression of RHEB mRNA in several different carcinoma histotypes, and an association between RHEB upregulation and poor prognosis in breast and head and neck cancers. To model gain of function in epithelial malignancy, we targeted Rheb expression to murine basal keratinocytes of transgenic mice at levels similar to those that occur in human squamous cancer cell lines. Juvenile transgenic epidermis displayed constitutive mTORC1 pathway activation, elevated cyclin D1 protein, and diffuse skin hyperplasia. Skin tumors subsequently developed with concomitant stromal angio-inflammatory foci, evidencing induction of an epidermal hypoxia-inducible factor-1 transcriptional program, and paracrine feed-forward activation of the interleukin-6-signal transducer and activator of transcription 3 pathway. Rheb-induced tumor persistence and neoplastic molecular alterations were mTORC1 dependent. Rheb markedly sensitized transgenic epidermis to squamous carcinoma induction following a single dose of Ras-activating carcinogen 7,12-dimethylbenz(a)anthracene. Our findings offer direct evidence that RHEB facilitates multistage carcinogenesis through induction of multiple oncogenic mechanisms, perhaps contributing to the poor prognosis of patients with cancers overexpressing RHEB.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Neuropéptidos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Neoplasias Cutáneas/metabolismo , Animales , Línea Celular Tumoral , Perfilación de la Expresión Génica , Humanos , Queratinocitos/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Transgénicos , Complejos Multiproteicos , Pronóstico , Proteínas , Proteína Homóloga de Ras Enriquecida en el Cerebro , Transducción de Señal , Serina-Treonina Quinasas TOR , Factores de Transcripción/metabolismo , Transgenes
18.
PLoS Genet ; 6(12): e1001258, 2010 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-21203491

RESUMEN

The transcription factor ATF2 has been shown to attenuate melanoma susceptibility to apoptosis and to promote its ability to form tumors in xenograft models. To directly assess ATF2's role in melanoma development, we crossed a mouse melanoma model (Nras(Q61K)::Ink4a⁻/⁻) with mice expressing a transcriptionally inactive form of ATF2 in melanocytes. In contrast to 7/21 of the Nras(Q61K)::Ink4a⁻/⁻ mice, only 1/21 mice expressing mutant ATF2 in melanocytes developed melanoma. Gene expression profiling identified higher MITF expression in primary melanocytes expressing transcriptionally inactive ATF2. MITF downregulation by ATF2 was confirmed in the skin of Atf2⁻/⁻ mice, in primary human melanocytes, and in 50% of human melanoma cell lines. Inhibition of MITF transcription by MITF was shown to be mediated by ATF2-JunB-dependent suppression of SOX10 transcription. Remarkably, oncogenic BRAF (V600E)-dependent focus formation of melanocytes on soft agar was inhibited by ATF2 knockdown and partially rescued upon shMITF co-expression. On melanoma tissue microarrays, a high nuclear ATF2 to MITF ratio in primary specimens was associated with metastatic disease and poor prognosis. Our findings establish the importance of transcriptionally active ATF2 in melanoma development through fine-tuning of MITF expression.


Asunto(s)
Factor de Transcripción Activador 2/metabolismo , Regulación Neoplásica de la Expresión Génica , Melanoma/metabolismo , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción Activador 2/genética , Animales , Línea Celular Tumoral , Células Cultivadas , Regulación hacia Abajo , Femenino , Humanos , Masculino , Melanocitos/metabolismo , Melanoma/genética , Melanoma/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción Asociado a Microftalmía/metabolismo , Piel/metabolismo , Piel/patología
19.
Clin Cancer Res ; 15(14): 4531-7, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19584170

RESUMEN

PURPOSE: The epidermal growth factor receptor (EGFR) is overexpressed in several tumor types, and its expression is influenced by the length of a 5'-end microsatellite repeat (CA)n: the longer the repeat, the lower the expression. Dinucleotide repeats accumulate insertion/deletion types of mutations in tumors with microsatellite instability. We designed this study to estimate the occurrence of these mutations in EGFR(CA)n and their relevance in carcinogenesis of microsatellite instability-positive colon and gastric tumors. EXPERIMENTAL DESIGN: We analyzed the frequency of EGFR(CA)n mutations in vivo in 55 colorectal and 14 gastric microsatellite instability-positive cancers, and in vitro in single-cell clone cultures of microsatellite instability-positive colon tumor cell line LS174. Single-cell clone cultures with different repeat lengths were analyzed by fluorescent-activated cell sorter for EGFR cell-surface expression. A correlation analysis was done between EGFR(CA)n mutations and mutations in KRAS, BRAF, and p53. RESULTS: Unlike single-cell clone cultures, which exhibited higher rate of deletions compared with insertions, most of EGFR(CA)n mutations in colon and gastric tumors were insertions. Longer EGFR(CA)n correlated with lower EGFR cell-surface expression in single-cell clone cultures. In colon cancers, the elongation of the repeat was associated negatively with mutations in KRAS and BRAF, but not in p53. CONCLUSIONS: The EGFR(CA)n elongation observed in tumors cannot be explained by an intrinsic property of this repeat favoring insertions versus deletions. Instead, a selection for repeat elongation occurs in microsatellite instability-positive tumors, leading to EGFR down-regulation. These findings suggest that in microsatellite instability-positive tumors current therapies targeting EGFR overexpression may have either no effect or an opposite to the expected effect.


Asunto(s)
Neoplasias del Colon/genética , Repeticiones de Dinucleótido/genética , Receptores ErbB/genética , Inestabilidad de Microsatélites , Regiones no Traducidas 5'/genética , Secuencia de Bases , Línea Celular Tumoral , Neoplasias del Colon/patología , Análisis Mutacional de ADN , Regulación hacia Abajo , Citometría de Flujo , Frecuencia de los Genes , Genes ras/genética , Genotipo , Humanos , Mutagénesis Insercional , Mutación , Poli A/genética , Reacción en Cadena de la Polimerasa , Polimorfismo Conformacional Retorcido-Simple , Proteínas Proto-Oncogénicas B-raf/genética , Eliminación de Secuencia , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Proteína p53 Supresora de Tumor/genética
20.
Methods Mol Biol ; 536: 463-72, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19378037

RESUMEN

The ''Rainbow western'' method permits detection of multiple antigens on a single protein blot. The procedure utilizes horseradish peroxidase (HRPase)-based detection with both a chemiluminescent and colorimetric substrate. In the ''Rainbow western'' procedure four different HRPase-colorimetric substrates that produce black, brown, red, and green colors are employed sequentially for detection and simultaneous display of four different antigens on the same blot. The Rainbow western methods have the potential to consolidate the work to analyze the expression levels of several proteins in studies of signaling pathways within biological samples. This technique could be particularly valuable for analysis of comigrating proteins, isoforms, and/or facilitating studies on phosphorylation, acetylation, and oligomerization of proteins tagged by the same epitope.


Asunto(s)
Antígenos/análisis , Western Blotting/métodos , Compuestos Cromogénicos/química , Coloración y Etiquetado/métodos , Animales , Western Blotting/instrumentación , Peroxidasa de Rábano Silvestre/metabolismo , Mediciones Luminiscentes , Proteínas Proto-Oncogénicas c-bcl-2/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...