Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Chem Soc Rev ; 47(14): 5312-5337, 2018 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-29770369

RESUMEN

The applications of nanoparticles (NPs) are increasing exponentially in consumer products, biotechnology and biomedicine, and humans, as well as the environment, are increasingly being exposed to NPs. Analogously, various (pathogenic) microorganisms are present at all the major exposure and entry sites for NPs in the human body as well as in environmental habitats. However, the field has just started to explore the complex interplay between NPs and microbes and the (patho)biological consequences. Based on recent insights, herein, we critically reviewed the available knowledge about the interaction of NPs with microbes and the analytical investigations including the latest intravital imaging tools. We have commented on how the NPs' characteristics influence complex formation with microorganisms, presented the underlying physicochemical forces, and provided examples of how this knowledge can be used to rationally control the NP-microbe interaction. We concluded by discussing the role of the biomolecule corona in NP-microbe crosstalk and speculated the impact of NP-microbe complex formation on the (patho)biological outcome and fate of microbial pathogens. The presented insights will not only support the field in engineering NPs with improved anti-microbial activity but also stimulate research on the biomedical and toxicological relevance of nanomaterial-microbiome complex formation for the anthropocene in general.


Asunto(s)
Bacterias Gramnegativas/metabolismo , Bacterias Grampositivas/metabolismo , Nanopartículas/metabolismo , Esporas Fúngicas/metabolismo , Animales , Antiinfecciosos/química , Antiinfecciosos/farmacología , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Grampositivas/efectos de los fármacos , Humanos , Microalgas/efectos de los fármacos , Microalgas/metabolismo , Nanopartículas/química , Nanopartículas/toxicidad , Imagen Óptica , Polen/efectos de los fármacos , Polen/metabolismo , Esporas Fúngicas/efectos de los fármacos
2.
Oncogene ; 35(26): 3351-64, 2016 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-26657154

RESUMEN

Proteolysis is not only a critical requirement for life, but the executing enzymes also play important roles in numerous pathological conditions, including cancer. Therefore, targeting proteases is clearly relevant for improving cancer patient care. However, to effectively control proteases, a profound knowledge of their mechanistic function as well as their regulation and downstream signalling in health and disease is required. The highly conserved protease Threonine Aspartase1 (Taspase1) is overexpressed in numerous liquid and solid malignancies and was characterized as a 'non-oncogene addiction' protease. Although Taspase1 was shown to cleave various regulatory proteins in humans as well as leukaemia provoking mixed lineage leukaemia fusions, our knowledge on its detailed functions and the underlying mechanisms contributing to cancer is still incomplete. Despite superficial similarity to type 2 asparaginases as well as Ntn proteases, such as the proteasome, Taspase1-related research so far gives us the picture of a unique protease exhibiting special features. Moreover, neither effective genetic nor chemical inhibitors for this enzyme are available so far, thus hampering not only to further dissect Taspase1's pathobiological functions but also precluding the assessment of its clinical impact. Based on recent insights, we here critically review the current knowledge of Taspase1's structure-function relationship and its mechanistic relevance for tumorigenesis obtained from in vitro and in vivo cancer models. We provide a comprehensive overview of tumour entities for which Taspase1 might be of predictive and therapeutic value, and present the respective experimental evidence. To stimulate progress in the field, a comprehensive overview of Taspase1 targeting approaches is presented, including coverage of Taspase1-related patents. We conclude by discussing future inhibition strategies and relevant challenges, which need to be resolved by the field.


Asunto(s)
Aspartato Amoníaco-Liasa/metabolismo , Endopeptidasas/metabolismo , Neoplasias/enzimología , Treonina/metabolismo , Investigación Biomédica Traslacional/métodos , Aspartato Amoníaco-Liasa/antagonistas & inhibidores , Aspartato Amoníaco-Liasa/genética , Endopeptidasas/genética , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/uso terapéutico , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Estructura Molecular , Neoplasias/genética , Neoplasias/prevención & control
3.
Chem Soc Rev ; 44(17): 6094-121, 2015 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-26065524

RESUMEN

Besides the wide use of engineered nanomaterials (NMs) in technical products, their applications are not only increasing in biotechnology and biomedicine, but also in the environmental field. While the physico-chemical properties and behaviour of NMs can be characterized accurately under idealized conditions, this is no longer the case in complex physiological or natural environments. Herein, proteins and other biomolecules rapidly bind to NMs, forming a protein/biomolecule corona that critically affects the NMs' (patho)biological and technical identities. As the corona impacts the in vitro and/or in vivo NM applications in humans and ecosystems, a mechanistic understanding of its relevance and of the biophysical forces regulating corona formation is mandatory. Based on recent insights, we here critically review and present an updated concept of corona formation and evolution. We comment on how corona signatures may be linked to effects at the nano-bio interface in physiological and environmental systems. In order to comprehensively analyse corona profiles and to mechanistically understand the coronas' biological/ecological impact, we present a tiered multidisciplinary approach. To stimulate progress in this field, we introduce the potential impact of the corona for NM-microbiome-(human)host interactions and the novel concept of 'nanologicals', i.e., the nanomaterial-specific targeting of molecular machines. We conclude by discussing the relevant challenges that still need to be resolved in this field.


Asunto(s)
Nanopartículas/química , Nanopartículas/metabolismo , Nanopartículas/toxicidad , Proteínas/metabolismo , Animales , Sangre/efectos de los fármacos , Técnicas de Química Analítica/métodos , Humanos , Concentración de Iones de Hidrógeno , Cinética , Pulmón/fisiología , Microbiota , Concentración Osmolar , Proteínas/química , Proteínas/farmacocinética , Pruebas de Toxicidad
4.
Leukemia ; 27(4): 792-802, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23038274

RESUMEN

The delicate balance between the synthesis and the degradation of proteins ensures cellular homeostasis. Proteases act in an irreversible manner and therefore have to be strictly regulated. The ubiquitin-proteasome system (UPS) is a major pathway for the proteolytic degradation of cellular proteins. As dysregulation of the UPS is observed in most cancers including leukemia, the UPS is a valid target for therapeutic intervention strategies. Ubiquitin-ligases selectively bind substrates to target them for poly-ubiquitinylation and proteasomal degradation. Therefore, pharmacological modulation of these proteins could allow a specific level of control. Increasing evidence accumulates that ubiquitin-ligases termed mammalian seven in absentia homologs (SIAHs) are not only critical for the pathogenesis of solid tumors but also for leukemogenesis. However, the relevance and therapeutic potential of SIAH-dependent processes has not been fully elucidated. Here, we summarize functions of SIAH ubiquitin-ligases in leukemias, how they select leukemia-relevant substrates for proteasomal degradation, and how the expression and activity of SIAH1 and SIAH2 can be modulated in vivo. We also discuss that epigenetic drugs belonging to the group of histone deacetylase inhibitors induce SIAH-dependent proteasomal degradation to accelerate the turnover of leukemogenic proteins. In addition, our review highlights potential areas for future research on SIAH proteins.


Asunto(s)
Leucemia/fisiopatología , Proteínas Nucleares/fisiología , Ubiquitina-Proteína Ligasas/fisiología , Animales , Regulación de la Expresión Génica , Humanos , Ratones , Mutación , Proteínas Nucleares/genética , Transducción de Señal , Especificidad por Sustrato , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/genética , Tirosina Quinasa 3 Similar a fms/genética
5.
Cell Death Dis ; 1: e51, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21364656

RESUMEN

Hearing impairment caused by ototoxic insults, such as noise or gentamicin is a worldwide health problem. As the molecular circuitries involved are not yet resolved, current otoprotective therapies are rather empirical than rational. Here, immunohistochemistry and western blotting showed that the cytoprotective protein survivin is expressed in the human and guinea pig cochlea. In the guinea pig model, moderate noise exposure causing only a temporary hearing impairment transiently evoked survivin expression in the spiral ligament, nerve fibers and the organ of Corti. Mechanistically, survivin upregulation may involve nitric oxide (NO)-induced Akt signaling, as enhanced expression of the endothelial NO synthase and phosphorylated Akt were detectable in some surviving-positive cell types. In contrast, intratympanic gentamicin injection inducing cell damage and permanent hearing loss correlated with attenuated survivin levels in the cochlea. Subsequently, the protective activity of the human and the guinea pig survivin orthologs against the ototoxin gentamicin was demonstrated by ectopic overexpression and RNAi-mediated depletion studies in auditory cells in vitro. These data suggest that survivin represents an innate cytoprotective resistor against stress conditions in the auditory system. The pharmacogenetic modulation of survivin may thus provide the conceptual basis for the rational design of novel therapeutic otoprotective strategies.


Asunto(s)
Cóclea/metabolismo , Proteínas Inhibidoras de la Apoptosis/fisiología , Animales , Células Cultivadas , Cóclea/citología , Femenino , Gentamicinas/toxicidad , Cobayas , Humanos , Proteínas Inhibidoras de la Apoptosis/genética , Proteínas Inhibidoras de la Apoptosis/metabolismo , Masculino , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Transducción de Señal , Survivin , Regulación hacia Arriba
6.
Oncogene ; 27(6): 732-40, 2008 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-17653085

RESUMEN

Therapy resistance represents a major problem for disease management in oncology. Histone deacetylase inhibitors (HDACi) have been shown to modulate the cell cycle, to induce apoptosis and to sensitize cancer cells for other chemotherapeutics. Our study shows that the HDACi valproic acid (VPA) and the ribonucleotide reductase inhibitor hydroxyurea (HU) potentiate the pro-apoptotic effects of each other towards several cancer cell lines. This correlates with the HU-induced degradation of the cyclin-dependent kinase inhibitors (CDKI) p21 and p27, mediated by the proteasome or caspase-3. Moreover, we found that caspase-3 activation is required for VPA-induced apoptosis. Remarkably, p21 and p27 can confer resistance against VPA and HU. Both CDKI interact with caspase-3 and compete with other caspase-3 substrates. Hence, p21 and p27 may contribute to chemotherapy resistance as apoptosis inhibitors. Since the biological effects of VPA and HU could be achieved at concentrations used in current treatment protocols, the combined application of these compounds might be considered as a potential strategy for cancer treatment.


Asunto(s)
Apoptosis , Ciclo Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Inhibidores de Histona Desacetilasas , Hidroxiurea/farmacología , Ribonucleótido Reductasas/farmacología , Caspasa 3/metabolismo , Inhibidores de Caspasas , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Humanos , Melanoma/enzimología , Ácido Valproico/farmacología
7.
Oncogene ; 27(13): 1853-64, 2008 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-17906697

RESUMEN

The combination of an increase in the cAMP-phosphodiesterase activity of h-prune and its interaction with nm23-H1 have been shown to be key steps in the induction of cellular motility in breast cancer cells. Here we present the molecular mechanisms of this interaction. The region of the nm23-h-prune interaction lies between S120 and S125 of nm23, where missense mutants show impaired binding; this region has been highly conserved throughout evolution, and can undergo serine phosphorylation by casein kinase I. Thus, the casein kinase I delta-epsilon specific inhibitor IC261 impairs the formation of the nm23-h-prune complex, which translates 'in vitro' into inhibition of cellular motility in a breast cancer cellular model. A competitive permeable peptide containing the region for phosphorylation by casein kinase I impairs cellular motility to the same extent as IC261. The identification of these two modes of inhibition of formation of the nm23-H1-h-prune protein complex pave the way toward new challenges, including translational studies using IC261 or this competitive peptide 'in vivo' to inhibit cellular motility induced by nm23-H1-h-prune complex formation during progression of breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas Portadoras/metabolismo , Movimiento Celular , Proteínas Inhibidoras de las Quinasas Dependientes de la Ciclina/metabolismo , Nucleósido Difosfato Quinasas NM23/metabolismo , Animales , Unión Competitiva , Neoplasias de la Mama/genética , Células COS , Caenorhabditis elegans/embriología , Caenorhabditis elegans/metabolismo , Proteínas Portadoras/genética , Comunicación Celular , Chlorocebus aethiops , Proteínas Inhibidoras de las Quinasas Dependientes de la Ciclina/genética , Drosophila melanogaster/embriología , Drosophila melanogaster/metabolismo , Humanos , Indoles/farmacología , Nucleósido Difosfato Quinasas NM23/genética , Fragmentos de Péptidos/farmacología , Floroglucinol/análogos & derivados , Floroglucinol/farmacología , Hidrolasas Diéster Fosfóricas/metabolismo , Monoéster Fosfórico Hidrolasas , Fosforilación , Xenopus laevis/embriología , Xenopus laevis/metabolismo
8.
J Pathol ; 211(5): 532-540, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17334981

RESUMEN

Survivin functions as an apoptosis inhibitor and a regulator of cell division in many tumours. The intracellular localization of survivin in tumours has been suggested as a prognostic marker. However, current reports are inconsistent and the underlying molecular mechanisms are not understood. The present study has examined the localization and prognostic value of nuclear and cytoplasmic survivin in the pre-therapeutic biopsies from 71 oral and oropharyngeal squamous carcinoma (OSCC) patients. Statistical analysis indicated that preferential nuclear versus cytoplasmic survivin correlated with favourable versus unfavourable disease outcome. Uni- and multi-variate analysis showed that in contrast to total survivin expression, the difference between nuclear and cytoplasmic survivin was a strong predictor for relapse-free survival (p=0.0003). As a potential underlying molecular mechanism, it is shown in OSCC cell lines that predominantly cytoplasmic survivin mediates protection against chemo- and radio-therapy-induced apoptosis. Importantly, the cytoplasmic localization of survivin is regulated by its nuclear export signal (NES), and export-deficient nuclear survivin is not cytoprotective. This study suggests that the difference between cytoplasmic and nuclear survivin is an indicator for survivin activity in tumour cells. Thus, this difference may serve as a predictive marker of outcome in OSCC patients undergoing multi-modality therapy. The pharmacogenetic interference with survivin's cytoplasmic localization is also to be pursued as a potential therapeutic strategy.


Asunto(s)
Biomarcadores de Tumor/análisis , Carcinoma de Células Escamosas/química , Inhibidores de Cisteína Proteinasa/análisis , Neoplasias de Cabeza y Cuello/química , Proteínas Asociadas a Microtúbulos/análisis , Proteínas de Neoplasias/análisis , Apoptosis/fisiología , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/radioterapia , Línea Celular Tumoral , Núcleo Celular/química , Citoplasma/química , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/radioterapia , Humanos , Inmunohistoquímica/métodos , Proteínas Inhibidoras de la Apoptosis , Estimación de Kaplan-Meier , Carioferinas/análisis , Neoplasias de la Boca/química , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias de la Boca/radioterapia , Señales de Exportación Nuclear/fisiología , Neoplasias Orofaríngeas/química , Neoplasias Orofaríngeas/tratamiento farmacológico , Neoplasias Orofaríngeas/radioterapia , Pronóstico , Receptores Citoplasmáticos y Nucleares/análisis , Survivin , Proteína Exportina 1
9.
Cell Death Differ ; 13(4): 576-85, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16179940

RESUMEN

The serine protease granzyme B (GrB) of cytotoxic lymphocytes efficiently induces apoptosis by direct activation of caspases and cleavage of central caspase substrates. We employed human GrB as an effector function in chimeric fusion proteins that also contain the EGFR ligand TGFalpha or an ErbB2-specific single-chain antibody fragment (scFv) for selective targeting to tumor cells. GrB-TGFalpha (GrB-T) and GrB-scFv(FRP5) (GrB-5) molecules expressed in the yeast Pichia pastoris were bifunctional, cleaving synthetic and natural GrB substrates, and binding specifically to cells expressing EGFR or ErbB2 target receptors. Upon cell binding the chimeric molecules were internalized into intracellular vesicles, but could be released into the cytosol by the endosomolytic reagent chloroquine. Treatment with picomolar to nanomolar concentrations of GrB-5 and GrB-T resulted in selective and rapid tumor cell killing, accompanied by clear signs of apoptosis such as chromatin condensation, membrane blebbing, formation of apoptotic bodies and activation of endogenous initiator and effector caspases.


Asunto(s)
Antineoplásicos/administración & dosificación , Apoptosis , Sistemas de Liberación de Medicamentos , Proteínas Recombinantes de Fusión/administración & dosificación , Serina Endopeptidasas/administración & dosificación , Anticuerpos/administración & dosificación , Anticuerpos/genética , Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Línea Celular Tumoral , Membrana Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Cloroquina/farmacología , Vesículas Citoplasmáticas/efectos de los fármacos , Vesículas Citoplasmáticas/enzimología , Relación Dosis-Respuesta a Droga , Endocitosis , Activación Enzimática/efectos de los fármacos , Receptores ErbB/metabolismo , Granzimas , Humanos , Concentración 50 Inhibidora , Pichia/genética , Pichia/metabolismo , Receptor ErbB-2/inmunología , Receptor ErbB-2/metabolismo , Proteínas Recombinantes de Fusión/biosíntesis , Serina Endopeptidasas/genética , Factor de Crecimiento Transformador alfa/administración & dosificación , Factor de Crecimiento Transformador alfa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...