Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
2.
Nanomaterials (Basel) ; 10(2)2020 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-32098406

RESUMEN

Nanomaterials have great potential for the prevention and treatment of cancer. Circulating tumor cells (CTCs) are cancer cells of solid tumor origin entering the peripheral blood after detachment from a primary tumor. The occurrence and circulation of CTCs are accepted as a prerequisite for the formation of metastases, which is the major cause of cancer-associated deaths. Due to their clinical significance CTCs are intensively discussed to be used as liquid biopsy for early diagnosis and prognosis of cancer. However, there are substantial challenges for the clinical use of CTCs based on their extreme rarity and heterogeneous biology. Therefore, methods for effective isolation and detection of CTCs are urgently needed. With the rapid development of nanotechnology and its wide applications in the biomedical field, researchers have designed various nano-sized systems with the capability of CTCs detection, isolation, and CTCs-targeted cancer therapy. In the present review, we summarize the underlying mechanisms of CTC-associated tumor metastasis, and give detailed information about the unique properties of CTCs that can be harnessed for their effective analytical detection and enrichment. Furthermore, we want to give an overview of representative nano-systems for CTC isolation, and highlight recent achievements in microfluidics and lab-on-a-chip technologies. We also emphasize the recent advances in nano-based CTCs-targeted cancer therapy. We conclude by critically discussing recent CTC-based nano-systems with high therapeutic and diagnostic potential as well as their biocompatibility as a practical example of applied nanotechnology.

3.
J Cancer Res Clin Oncol ; 146(2): 343-356, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31932908

RESUMEN

PURPOSE: We set out to determine whether clinically tested epigenetic drugs against class I histone deacetylases (HDACs) affect hallmarks of the metastatic process. METHODS: We treated permanent and primary renal, lung, and breast cancer cells with the class I histone deacetylase inhibitors (HDACi) entinostat (MS-275) and valproic acid (VPA), the replicative stress inducer hydroxyurea (HU), the DNA-damaging agent cis-platinum (L-OHP), and the cytokine transforming growth factor-ß (TGFß). We used proteomics, quantitative PCR, immunoblot, single cell DNA damage assays, and flow cytometry to analyze cell fate after drug exposure. RESULTS: We show that HDACi interfere with DNA repair protein expression and trigger DNA damage and apoptosis alone and in combination with established chemotherapeutics. Furthermore, HDACi disrupt the balance of cell adhesion protein expression and abrogate TGFß-induced cellular plasticity of transformed cells. CONCLUSION: HDACi suppress the epithelial-mesenchymal transition (EMT) and compromise the DNA integrity of cancer cells. These data encourage further testing of HDACi against tumor cells.


Asunto(s)
Reparación del ADN/fisiología , Proteínas de Unión al ADN/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Neoplasias/tratamiento farmacológico , Animales , Benzamidas/farmacología , Plasticidad de la Célula/efectos de los fármacos , Cisplatino/farmacología , Enzimas Reparadoras del ADN/metabolismo , Resistencia a Antineoplásicos , Humanos , Hidroxiurea/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Metástasis de la Neoplasia , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Piridinas/farmacología , Factor de Crecimiento Transformador beta/farmacología , Ácido Valproico/farmacología
4.
J Histochem Cytochem ; 67(11): 845-855, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31510846

RESUMEN

Constitutively expressed endothelial nitric oxide synthase (eNOS) is supposed to play a role in noise-induced nitric oxide (NO)-production. It is commonly known that intense noise exposure results in inducible NOS (iNOS) expression and increased NO-production, but knowledge about a contribution of the eNOS isoform is still lacking. Effects of noise exposure on eNOS immunolabeling were determined in male guinea pigs (n=24). For light microscopic analysis, 11 animals were exposed to 90 dB for 1 hr and 6 animals were used as controls. After exposure, eNOS immunostaining was performed on paraffin sections, and the staining intensities were quantified for 4 cochlear regions. For electron microscopic analysis, 2 animals were exposed for 2 hr to 90 dB and 5 animals were used as controls. The intensity of eNOS immunolabeling was found to be already comprehensively increased 1 hr after noise exposure to 90 dB. At the ultrastructural level, a clear increase in eNOS immunolabeling was found in microtubules-rich areas of cochlear cuticular structures. Hence, our findings indicate that the reticular lamina forming the endolymph-perilymph barrier at the apical side of the organ of Corti is involved in a fast intrinsic otoprotective mechanism of the cochlea.


Asunto(s)
Cóclea/metabolismo , Cobayas/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Ruido/efectos adversos , Animales , Cóclea/ultraestructura , Pérdida Auditiva Provocada por Ruido/metabolismo , Inmunohistoquímica , Masculino , Óxido Nítrico Sintasa de Tipo III/análisis
5.
Int J Nanomedicine ; 14: 4187-4209, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31289440

RESUMEN

Circulating tumor cells (CTCs) are disseminated cancer cells. The occurrence and circulation of CTCs seem key for metastasis, still the major cause of cancer-associated deaths. As such, CTCs are investigated as predictive biomarkers. However, due to their rarity and heterogeneous biology, CTCs' practical use has not made it into the clinical routine. Clearly, methods for the effective isolation and reliable detection of CTCs are urgently needed. With the development of nanotechnology, various nanosystems for CTC isolation and enrichment and CTC-targeted cancer therapy have been designed. Here, we summarize the relationship between CTCs and tumor metastasis, and describe CTCs' unique properties hampering their effective enrichment. We comment on nanotechnology-based systems for CTC isolation and recent achievements in microfluidics and lab-on-a-chip technologies. We discuss recent advances in CTC-targeted cancer therapy exploiting the unique properties of nanomaterials. We conclude by introducing developments in CTC-directed nanosystems and other advanced technologies currently in (pre)clinical research.


Asunto(s)
Biomarcadores de Tumor/análisis , Separación Celular/métodos , Nanomedicina/métodos , Células Neoplásicas Circulantes/patología , Biomarcadores de Tumor/aislamiento & purificación , Materiales Biomiméticos , Grafito , Humanos , Dispositivos Laboratorio en un Chip , Microfluídica/instrumentación , Microfluídica/métodos , Nanoestructuras/química , Nanotecnología/métodos , Nanotubos de Carbono
7.
Eur J Surg Oncol ; 45(6): 1046-1053, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30857877

RESUMEN

INTRODUCTION: Recognizing the prognostic power differentiating HPV-associated oropharyngeal squamous cell cancer (OPSCC) from OPSCC with other causes, the UICC Cancer Staging Manual 8th edition realizes significant changes from the 7th edition. Purpose of this study was to evaluate the differences of prognostic impact between the 7th and the latest edition of TNM Classification as well as to examine risk factors like extranodal extension (ENE) and lymph node ratio (LNR) for HPV-mediated OPSCC. MATERIAL AND METHODS: The study includes 255 patients with OPSCC and initial diagnosis between 2008 and 2015. HPV status was determined according to p16 immunohistochemistry (IHC) and all patients were classified as defined by 7th and 8th edition of UICC. Prognostic influence of ENE and LNR was analyzed for patients with HPV-mediated OPSCC. RESULTS: 41.2% of the OPSCC were p16-positive. Implementation of the 8th edition of the UICC lead to a better differentiation between the respective stages. Regarding HPV-positive OPSCC, Kaplan-Meier survival curves showed a significantly better overall survival (OS) for patients with a LNR ≤10% as well as for patients with negative ENE status (p = 0.004, p = 0.008). CONCLUSION: 8th edition of UICC achieves to differentiate properly between the UICC stages. However, the staging rule of ignoring ENE in HPV-mediated OPSCC should be further analyzed. Moreover LNR might be a possible additional prognostic factor - especially regarding HPV-positive tumors.


Asunto(s)
Neoplasias de Cabeza y Cuello/diagnóstico , Estadificación de Neoplasias/métodos , Neoplasias Orofaríngeas/diagnóstico , Carcinoma de Células Escamosas de Cabeza y Cuello/diagnóstico , Femenino , Estudios de Seguimiento , Alemania/epidemiología , Neoplasias de Cabeza y Cuello/mortalidad , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Neoplasias Orofaríngeas/mortalidad , Pronóstico , Estudios Retrospectivos , Carcinoma de Células Escamosas de Cabeza y Cuello/mortalidad , Tasa de Supervivencia/tendencias
8.
Biol Chem ; 400(11): 1465-1479, 2019 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-30903749

RESUMEN

The establishment of novel biomarkers in liquid biopsies of cancer patients has come more into focus in prognostic and diagnostic research efforts. Due to their prognostic relevance disseminated tumor cells or circulating tumor cells are the subject of intensive research and are discussed as early diagnostic indicators for treatment failure and the formation of micrometastases. A potential association of this early-systemic tumor component with poor prognosis of cancer patients could be already demonstrated for various entities including breast, colon, lung, melanoma, ovarian and prostate cancers. Thus, the detection of circulating tumor cells seems to be also applicable for minimal-invasive monitoring of therapy progress in head and neck cancer patients. A major problem of the use in clinical routine is that circulating tumor cells could not be detected by modern imaging techniques. To overcome these limitations highly sensitive detection methods and techniques for their molecular characterization are urgently needed allowing mechanistic understanding and targeting of circulating tumor cells. Especially the medical application of nanotechnology (nanomedical methods) has made valuable contributions to the field. Here, we want to provide a comprehensive overview on (nanomedical) detection methods for circulating tumor cells and discuss their merits, pitfalls and future perspectives especially for head and neck solid squamous cell carcinoma (HNSCC) patients.


Asunto(s)
Carcinoma de Células Escamosas/diagnóstico , Neoplasias de Cabeza y Cuello/diagnóstico , Nanomedicina , Células Neoplásicas Circulantes/patología , Humanos
9.
Biomaterials ; 192: 551-559, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30530244

RESUMEN

Multidrug-resistant bacterial infections are a global health threat. Nanoparticles are thus investigated as novel antibacterial agents for clinical practice, including wound dressings and implants. We report that nanoparticles' bactericidal activity strongly depends on their physical binding to pathogens, including multidrug-resistant primary clinical isolates, such as Staphylococcus aureus, Klebsiella pneumoniae or Enterococcus faecalis. Using controllable nanoparticle models, we found that nanoparticle-pathogen complex formation was enhanced by small nanoparticle size rather than material or charge, and was prevented by 'stealth' modifications. Nanoparticles seem to preferentially bind to Gram-positive pathogens, such as Listeria monocytogenes, S. aureus or Streptococcus pyrogenes, correlating with enhanced antibacterial activity. Bacterial resistance to metal-based nanoparticles was mediated by biomolecule coronas acquired in pathophysiological environments, such as wounds, the lung, or the blood system. Biomolecule corona formation reduced nanoparticles' binding to pathogens, but did not impact nanoparticle dissolution. Our results provide a mechanistic explanation why nano-sized antibiotics may show reduced activity in clinically relevant environments, and may inspire future nanoantibiotic designs with improved and potentially pathogen-specific activity.


Asunto(s)
Antibacterianos/farmacología , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Viabilidad Microbiana/efectos de los fármacos , Nanopartículas/química , Adsorción , Escherichia coli/efectos de los fármacos , Escherichia coli/ultraestructura , Pruebas de Sensibilidad Microbiana , Nanopartículas/ultraestructura
10.
Sci Rep ; 8(1): 7326, 2018 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-29743718

RESUMEN

Head and neck squamous cell carcinoma (HNSCC) often metastasize to lymph nodes resulting in poor prognosis for patients. Unfortunately, the underlying molecular mechanisms contributing to tumour aggressiveness, recurrences, and metastasis are still not fully understood. However, such knowledge is key to identify biomarkers and drug targets to improve prognosis and treatments. Consequently, we performed genome-wide expression profiling of 15 primary HNSSCs compared to corresponding lymph node metastases and non-malignant tissue of the same patient. Differentially expressed genes were bioinformatically exploited applying stringent filter criteria, allowing the discrimination between normal mucosa, primary tumours, and metastases. Signalling networks involved in invasion contain remodelling of the extracellular matrix, hypoxia-induced transcriptional modulation, and the recruitment of cancer associated fibroblasts, ultimately converging into a broad activation of PI3K/AKT-signalling pathway in lymph node metastasis. Notably, when we compared the diagnostic and prognostic value of sequencing data with our expression analysis significant differences were uncovered concerning the expression of the receptor tyrosine kinases EGFR and ERBB2, as well as other oncogenic regulators. Particularly, upregulated receptor tyrosine kinase combinations for individual patients varied, implying potential compensatory and resistance mechanisms against specific targeted therapies. Collectively, we here provide unique transcriptional profiles for disease predictions and comprehensively analyse involved signalling pathways in advanced HNSCC.


Asunto(s)
Perfilación de la Expresión Génica , Transducción de Señal/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Femenino , Humanos , Metástasis Linfática , Masculino , Persona de Mediana Edad , Pronóstico , Proteínas Tirosina Quinasas Receptoras/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/diagnóstico
11.
J Mol Cell Biol ; 10(1): 33-47, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28992066

RESUMEN

Transcription factor TFIIA is controlled by complex regulatory networks including proteolysis by the protease Taspase 1, though the full impact of cleavage remains elusive. Here, we demonstrate that in contrast to the general assumption, de novo produced TFIIA is rapidly confined to the cytoplasm via an evolutionary conserved nuclear export signal (NES, amino acids 21VINDVRDIFL30), interacting with the nuclear export receptor Exportin-1/chromosomal region maintenance 1 (Crm1). Chemical export inhibition or genetic inactivation of the NES not only promotes TFIIA's nuclear localization but also affects its transcriptional activity. Notably, Taspase 1 processing promotes TFIIA's nuclear accumulation by NES masking, and modulates its transcriptional activity. Moreover, TFIIA complex formation with the TATA box binding protein (TBP) is cooperatively enhanced by inhibition of proteolysis and nuclear export, leading to an increase of the cell cycle inhibitor p16INK, which is counteracted by prevention of TBP binding. We here identified a novel mechanism how proteolysis and nuclear transport cooperatively fine-tune transcriptional programs.


Asunto(s)
Núcleo Celular/metabolismo , Endopeptidasas/metabolismo , Carioferinas/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Factor de Transcripción TFIIA/metabolismo , Transporte Activo de Núcleo Celular , Línea Celular , Células HeLa , Humanos , Modelos Moleculares , Señales de Exportación Nuclear , Conformación Proteica , Factor de Transcripción TFIIA/análisis , Factor de Transcripción TFIIA/genética , Activación Transcripcional , Proteína Exportina 1
13.
FASEB J ; 30(3): 1012-22, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26578689

RESUMEN

From the beginning of life, proteases are key to organismal development comprising morphogenesis, cellular differentiation, and cell growth. Regulated proteolytic activity is essential for the orchestration of multiple developmental pathways, and defects in protease activity can account for multiple disease patterns. The highly conserved protease threonine aspartase 1 is a member of such developmental proteases and critically involved in the regulation of complex processes, including segmental identity, head morphogenesis, spermatogenesis, and proliferation. Additionally, threonine aspartase 1 is overexpressed in numerous liquid as well as in solid malignancies. Although threonine aspartase 1 is able to cleave the master regulator mixed lineage leukemia protein as well as other regulatory proteins in humans, our knowledge of its detailed pathobiological function and the underlying molecular mechanisms contributing to development and disease is still incomplete. Moreover, neither effective genetic nor chemical inhibitors for this enzyme are available so far precluding the detailed dissection of the pathobiological functions of threonine aspartase 1. Here, we review the current knowledge of the structure-function relationship of threonine aspartase 1 and its mechanistic impact on substrate-mediated coordination of the cell cycle and development. We discuss threonine aspartase 1-mediated effects on cellular transformation and conclude by presenting a short overview of recent interference strategies.


Asunto(s)
Endopeptidasas/metabolismo , Péptido Hidrolasas/metabolismo , Animales , Ciclo Celular/fisiología , Humanos
14.
Gerontology ; 62(2): 173-81, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26159786

RESUMEN

Survivin (BIRC5) is highly expressed in the vast majority of human cancers and is associated with chemotherapy resistance, increased tumor recurrence and shortened patient survival, making it an attractive therapeutic target. Initially identified as an inhibitor of apoptosis protein, it also plays a major role in the regulation of cell division. As such, it acts as a subunit of the chromosomal passenger complex, composed of the mitotic kinase aurora B, borealin and inner centromere protein, and is essential for proper chromosome segregation and cytokinesis. For both biological functions, interaction of survivin's nuclear export signal with the nuclear export receptor chromosome region maintenance 1 is absolutely essential. The timely orchestration of survivin's wide protein interaction repertoire is further modulated by different posttranslational modifications occurring in a cell-cycle-dependent manner. Recent data furthermore indicate additional roles of survivin in the DNA damage response, contributing to therapy resistance, yet the underlying molecular details are still not completely resolved. This also holds true for a potential involvement of survivin in senescence regulation. An age-related accumulation of survivin probably contributes to the apoptosis resistance observed in aged as well as in senescent cells, while it might promote escape from therapy-induced senescence. This review seeks to integrate the current knowledge on survivin's diverse and complex biological functions. By linking the 'old' facts about survivin with recent findings in research areas such as DNA damage response and aging, we want to highlight survivin's crucial role in a variety of cellular processes.


Asunto(s)
Envejecimiento , Apoptosis , Senescencia Celular , Reparación del ADN , Proteínas Inhibidoras de la Apoptosis/metabolismo , Mitosis , Neoplasias/metabolismo , Humanos , Survivin
15.
Biol Chem ; 396(4): 367-76, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25720119

RESUMEN

Proteases are key regulators of life. Human Threonine Aspartase1 processes substrates, such as the mixed-lineage leukemia (MLL) protein, containing two cleavage sites, CS1 and CS2. Likewise, MLL's Drosophila ortholog trithorax is cleaved by Drosophila Threonine Aspartase1 (dTasp), suggesting a mechanistic coevolution. However, a detailed analysis of dTasp's function was missing so far. Here, active and inactive dTasp mutants allowed to compare substrate recognition and cleavage site selectivity of human and Drosophila enzymes. In contrast to the human protease, our cell-based assay revealed a preferential processing of CS2-like (QLD↓Gx[xD/Dx]) targets for dTasp, whereas cleavage of CS1-like targets (QVD↓Gx[xD/Dx]) was significantly impaired. Systematic mutagenesis of the CS2 sequence defined the motif x[FILMW]D↓Gx[xD/Dx] as the consensus cleavage sequence for dTasp. Substrate species selectivity of the enzymes was uncovered by demonstrating that dTasp cleaves Drosophila TFIIA, but not the human ortholog, suggesting evolutionary divergence of TFIIA downstream networks. Also, Drosophila USF2 was neither predicted nor cleaved by dTasp. Moreover, we found that dTasp cleavage site selectivity is independent of heterocomplex formation, as dTasp exists predominantly as an αß-monomer. Collectively, we provide novel insights into evolutionary similarities and divergence concerning Threonine Aspartase1 function in different species, which may aid to dissect and better target human Threonine Aspartase1 in malignancies.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/enzimología , Endopeptidasas/metabolismo , Secuencia de Aminoácidos , Animales , Drosophila/química , Drosophila/metabolismo , Proteínas de Drosophila/química , Endopeptidasas/química , Células HeLa , Humanos , Datos de Secuencia Molecular , Multimerización de Proteína , Especificidad de la Especie , Especificidad por Sustrato , Factor de Transcripción TFIIA/metabolismo , Factores Estimuladores hacia 5'/metabolismo
16.
FASEB J ; 29(5): 1973-85, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25634959

RESUMEN

Human Taspase1 is essential for development and cancer by processing critical regulators, such as the mixed-lineage leukemia protein. Likewise, its ortholog, trithorax, is cleaved by Drosophila Taspase1 (dTaspase1), implementing a functional coevolution. To uncover novel mechanism regulating protease function, we performed a functional analysis of dTaspase1 and its comparison to the human ortholog. dTaspase1 contains an essential nucleophile threonine(195), catalyzing cis cleavage into its α- and ß-subunits. A cell-based assay combined with alanine scanning mutagenesis demonstrated that the target cleavage motif for dTaspase1 (Q(3)[F/I/L/M](2)D(1)↓G(1')X(2')X(3')) differs significantly from the human ortholog (Q(3)[F,I,L,V](2)D(1)↓G(1')x(2')D(3')D(4')), predicting an enlarged degradome containing 70 substrates for Drosophila. In contrast to human Taspase1, dTaspase1 shows no discrete localization to the nucleus/nucleolus due to the lack of the importin-α/nucleophosmin1 interaction domain (NoLS) conserved in all vertebrates. Consequently, dTaspase1 interacts with neither the Drosophila nucleoplasmin-like protein nor human nucleophosmin1. The impact of localization on the protease's degradome was confirmed by demonstrating that dTaspase1 did not efficiently process nuclear substrates, such as upstream stimulatory factor 2. However, genetic introduction of the NoLS into dTaspase1 restored its nucleolar localization, nucleophosmin1 interaction, and efficient cleavage of nuclear substrates. We report that evolutionary functional divergence separating vertebrates from invertebrates can be achieved for proteases by a transport/localization-regulated mechanism.


Asunto(s)
Evolución Biológica , Nucléolo Celular/metabolismo , Núcleo Celular/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Endopeptidasas/metabolismo , Péptido Hidrolasas/metabolismo , Secuencia de Aminoácidos , Animales , Western Blotting , Células Cultivadas , Drosophila/crecimiento & desarrollo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Masculino , Microscopía Confocal , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Filogenia , Transporte de Proteínas , Proteolisis , Homología de Secuencia de Aminoácido , Transducción de Señal
17.
Nat Protoc ; 9(9): 2030-44, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25079427

RESUMEN

Nanoparticle applications in biotechnology and biomedicine are steadily increasing. In biological fluids, proteins bind to nanoparticles that form the protein corona, crucially affecting the nanoparticles' biological identity. As the corona affects in vitro and/or in vivo nanoparticle applications, we developed a method to obtain time-resolved protein corona profiles formed on various nanoparticles. After incubation in plasma or a similar biofluid, or after injection into a mouse, the first analytical step is sedimentation of the nanoparticle-protein complexes through a sucrose cushion, thereby allowing analysis of early corona formation time points. Next, corona profiles are visualized by gel electrophoresis and quantitatively analyzed after tryptic digestion using label-free liquid chromatography-high-resolution mass spectrometry. In contrast to other approaches, our established methodology allows the researcher to obtain qualitative and quantitative high-resolution corona signatures. The protocol can be readily extended to the investigation of protein coronas from various nanomaterials (as an example, we applied this protocol to different silica nanoparticles (SiNPs) and polystyrene nanoparticles (PSNPs)). Depending on the number of samples, the protocol from nanoparticle-protein complex recovery to data evaluation takes ~8-12 d to complete.


Asunto(s)
Fraccionamiento Químico/métodos , Técnicas de Química Analítica/métodos , Nanopartículas/química , Proteínas/análisis , Proteínas/aislamiento & purificación , Animales , Cromatografía Liquida , Electroforesis , Espectrometría de Masas , Ratones , Poliestirenos , Dióxido de Silicio , Sacarosa , Tripsina
18.
FASEB J ; 26(8): 3421-9, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22623586

RESUMEN

Taspase1 mediates cleavage of the mixed lineage leukemia (MLL) protein and leukemia-provoking MLL fusions and promotes solid malignancies. Currently, no effective and specific Taspase1 inhibitors are available, precluding its therapeutic exploitation. As the Taspase1 proenzyme is autoproteolytically cleaved and assumed to assemble into an active αßßα heterodimer, we attempted to interfere with its activity by targeting Taspase1's dimerization. Notably, enforced expression of inactive Taspase1 mutants, aiming to inhibit formation of active protease dimers, was not inhibitory. Immunoprecipitation, gel filtration, and in vivo protein interaction assays revealed that active Taspase1 exists predominantly as an αß monomer in living cells, providing an explanation why overexpression of inactive mutants was not trans-dominant. To alternatively test the biological consequences of enforced dimerization, we engineered Taspase1 variants containing the Jun/Fos dimerization motif. In absence of the respective interaction partners, the protease fusions were fully active, while enforcing dimerization by coexpression significantly inhibited processing of several target proteins in living cells. Our study provides the first evidence that Taspase1 is already active as an αß monomer, arguing against heterocomplex formation being required for its pathobiological activity. Thus, it clearly supports strategies aiming to inhibit the cancer-promoting activity of Taspase1 by the identification of chemical decoys enforcing its dimerization.


Asunto(s)
Endopeptidasas/efectos de los fármacos , Regulación Alostérica , Línea Celular Tumoral , Endopeptidasas/genética , Humanos , Inhibidores de Proteasas/farmacología , Ingeniería de Proteínas , Multimerización de Proteína/efectos de los fármacos
19.
PLoS One ; 7(5): e34142, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22570686

RESUMEN

BACKGROUND: The chromosomal translocation t(4;11)(q21;q23) is associated with high-risk acute lymphoblastic leukemia of infants. The resulting AF4•MLL oncoprotein becomes activated by Taspase1 hydrolysis and is considered to promote oncogenic transcriptional activation. Hence, Taspase1's proteolytic activity is a critical step in AF4•MLL pathophysiology. The Taspase1 proenzyme is autoproteolytically processed in its subunits and is assumed to assemble into an αßßα-heterodimer, the active protease. Therefore, we investigated here whether overexpression of catalytically inactive Taspase1 variants are able to interfere with the proteolytic activity of the wild type enzyme in AF4•MLL model systems. METHODOLOGY/FINDINGS: The consequences of overexpressing the catalytically dead Taspase1 mutant, Taspase1(T234V), or the highly attenuated variant, Taspase1(D233A), on Taspase1's processing of AF4•MLL and of other Taspase1 targets was analyzed in living cancer cells employing an optimized cell-based assay. Notably, even a nine-fold overexpression of the respective Taspase1 mutants neither inhibited Taspase1's cis- nor trans-cleavage activity in vivo. Likewise, enforced expression of the α- or ß-subunits showed no trans-dominant effect against the ectopically or endogenously expressed enzyme. Notably, co-expression of the individual α- and ß-subunits did not result in their assembly into an enzymatically active protease complex. Probing Taspase1 multimerization in living cells by a translocation-based protein interaction assay as well as by biochemical methods indicated that the inactive Taspase1 failed to assemble into stable heterocomplexes with the wild type enzyme. CONCLUSIONS: Collectively, our results demonstrate that inefficient heterodimerization appears to be the mechanism by which inactive Taspase1 variants fail to inhibit wild type Taspase1's activity in trans. Our work favours strategies targeting Taspase1's catalytic activity rather than attempts to block the formation of active Taspase1 dimers to interfere with the pathobiological function of AF4•MLL.


Asunto(s)
Endopeptidasas/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Translocación Genética , Secuencia de Aminoácidos , Línea Celular Tumoral , Cromosomas Humanos Par 11 , Cromosomas Humanos Par 4 , Endopeptidasas/metabolismo , Activación Enzimática/genética , Expresión Génica , Variación Genética , Humanos , Lactante , Modelos Biológicos , Complejos Multiproteicos , Mutación , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Mapeo de Interacción de Proteínas/métodos , Multimerización de Proteína
20.
Gene ; 469(1-2): 9-17, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-20727954

RESUMEN

BACKGROUND: The guinea pig is widely used as a model to study (patho)physiological processes, such as neurodegenerative disorders. Survivin's dual function as an apoptosis inhibitor and a mitotic regulator is crucial not only for ordered development but its modulation seems crucial also under disease conditions. However, data on the expression and function of the guinea pig Survivin protein (Survivin(Gp)) are currently lacking. RESULTS: Here, we here report the cloning and functional characterization of Survivin(Gp). The respective cDNA was cloned from spleen mRNA, containing a 426 bp open reading frame encoding for a protein of 142aa. Survivin(Gp) displays a high homology to the human and murine orthologue, especially in domains critical for function, such as binding sites for chromosomal passenger complex (CPC) proteins and the nuclear export signal (NES). Notably, phylogenetic analyses revealed that Survivin(Gp) is more related to humans than to rodents. Ectopic expression studies of a Survivin(Gp)-GFP fusion confirmed its dynamic intracellular localization, analogous to the human and murine counterparts. In interphase cells, Survivin(Gp)-GFP was predominantly cytoplasmic and accumulated in the nucleus following export inhibition with leptomycin B (LMB). A typical CPC protein localization during mitosis was observed for Survivin(Gp)-GFP. Microinjection experiments together with genetic knockout demonstrated that the NES is essential for the anti-apoptotic and regulatory role of Survivin(Gp) during cell division. In vivo protein interaction assays further demonstrated its dimerization with human Survivin and its interaction with human CPC proteins. Importantly, RNAi-depletion studies show that Survivin(Gp) can fully substitute for human Survivin as an apoptosis inhibitor and a mitotic effector. Immunohistochemistry, immunofluorescence, and western blotting were employed to detect Survivin expression in guinea pig tissues. Besides its expression in proliferating tissues, such as spleen and liver, we also found Survivin in terminally differentiated cell types. Importantly, Survivin was detectable also in the cochlea, suggesting a potential role for Survivin in the auditory system. CONCLUSIONS: We provide the first experimental evidence for the expression of Survivin in the guinea pig. As Survivin(Gp) can substitute for known functions of human Survivin, the guinea pig model will now also allow investigating Survivin's (patho)physiological role and to test Survivin-directed potential therapeutic strategies.


Asunto(s)
Cobayas/genética , Proteínas Inhibidoras de la Apoptosis/genética , Secuencia de Aminoácidos , Animales , Clonación Molecular , Oído Interno/metabolismo , Proteínas Asociadas a Microtúbulos , Mitosis , Modelos Animales , Datos de Secuencia Molecular , Filogenia , Homología de Secuencia de Aminoácido , Survivin , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...