Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Neurochem ; 148(2): 168-187, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30144068

RESUMEN

S100B is a Ca2+ -binding protein mainly concentrated in astrocytes. Its levels in biological fluids (cerebrospinal fluid, peripheral and cord blood, urine, saliva, amniotic fluid) are recognized as a reliable biomarker of active neural distress. Although the wide spectrum of diseases in which the protein is involved (acute brain injury, neurodegenerative diseases, congenital/perinatal disorders, psychiatric disorders) reduces its specificity, its levels remain an important aid in monitoring the trend of the disorder. Mounting evidence now points to S100B as a Damage-Associated Molecular Pattern molecule which, when released at high concentration, through its Receptor for Advanced Glycation Endproducts, triggers tissue reaction to damage in a series of different neural disorders. This review addresses this novel scenario, presenting data indicating that S100B levels and/or distribution in the nervous tissue of patients and/or experimental models of different neural disorders, for which the protein is used as a biomarker, are directly related to the progress of the disease: acute brain injury (ischemic/hemorrhagic stroke, traumatic injury), neurodegenerative diseases (Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, multiple sclerosis), congenital/perinatal disorders (Down syndrome, spinocerebellar ataxia-1), psychiatric disorders (schizophrenia, mood disorders), inflammatory bowel disease. In many cases, over-expression/administration of the protein induces worsening of the disease, whereas its deletion/inactivation produces amelioration. This review points out that the pivotal role of the protein resulting from these data, opens the perspective that S100B may be regarded as a therapeutic target for these different diseases, which appear to share some common features reasonably attributable to neuroinflammation, regardless their origin.


Asunto(s)
Biomarcadores , Enfermedades del Sistema Nervioso , Subunidad beta de la Proteína de Unión al Calcio S100 , Animales , Humanos
2.
J Cell Physiol ; 231(10): 2081-7, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-26791139

RESUMEN

Currently, there is much interest in the characterization of metabolic profiling of cancer stem cells (CSCs), a small subset of tumor cells with self-renewal capacity. Indeed, ever-growing evidence indicate that metabolism and stemness are highly intertwined processes in tumor tissue. In this review, we analyze the potential metabolic targeting strategies for eradicating CSCs that could help to develop a more effective therapeutic approach for gastrointestinal cancers. Indeed, the successful elimination of a tumor requires an anticancer therapy that affects both cancer cells and CSCs. The observation that gastrointestinal CSCs possess higher inducible nitric oxide sinthase (iNOS) expression, lower reactive oxygen species (ROS) production, and a different metabolism respect to no-CSCs tumor cells has paved the way to develop drugs targeting CSC specific signaling. In particular, several studies have highlighted that metformin, aldehyde dehydrogenase 1, and iNOS inhibitors selectively suppressed CSC growth and that combinatorial therapy of them with standard chemotherapeutic drugs had a synergistic effect resulting in reduced tumor burden and delayed tumor recurrence. Thus, the possibility of combining specific CSC metabolism inhibitors with existing therapeutic approaches could have profound anticancer effects, changing the conventional treatment approaches to gastrointestinal cancers. J. Cell. Physiol. 231: 2081-2087, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias Gastrointestinales/metabolismo , Células Madre Neoplásicas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Línea Celular Tumoral , Resistencia a Antineoplásicos/fisiología , Neoplasias Gastrointestinales/tratamiento farmacológico , Humanos , Células Madre Neoplásicas/efectos de los fármacos
3.
J Pathol ; 236(4): 479-90, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25875314

RESUMEN

Chronic inflammation is a leading cause of neoplastic transformation in many human cancers and especially in colon cancer (CC), in part due to tumour promotion by nitric oxide (NO) generated at inflammatory sites. It has also been suggested that high NO synthesis, secondary to inducible NO synthase (iNOS) expression, is a distinctive feature of cancer stem cells (CSCs), a small subset of tumour cells with self-renewal capacity. In this study we explored the contribution of NO to the development of colon CSC features and evaluated potential strategies to treat CC by modulating NO production. Our data show an integral role for endogenous NO and iNOS activity in the biology of colon CSCs. Indeed, colon CSCs with high endogenous NO production (NO(high)) displayed higher tumourigenic abilities than NO(low) fractions. The blockade of endogenous NO availability, using either a specific iNOS inhibitor or a genetic knock-down of iNOS, resulted in a significant reduction of colon CSC tumourigenic capacities in vitro and in vivo. Interestingly, analysis of genes altered by iNOS-directed shRNA showed that the knockdown of iNOS expression was associated with a significant down-regulation of signalling pathways involved in stemness and tumour progression in colon CSCs. These findings confirm that endogenous NO plays an important role in defining the stemness properties of colon CSCs through cross-regulation of several cellular signalling pathways. This discovery could shed light on the mechanisms by which NO induces the growth and invasiveness of CC, providing new insights into the link between inflammation and colon tumourigenesis.


Asunto(s)
Neoplasias Colorrectales/enzimología , Células Madre Neoplásicas/enzimología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico/metabolismo , Antígeno AC133 , Animales , Antígenos CD/metabolismo , Antineoplásicos/farmacología , Biomarcadores de Tumor/metabolismo , Células CACO-2 , Proliferación Celular , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Inhibidores Enzimáticos/farmacología , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Glicoproteínas/metabolismo , Células HCT116 , Células HEK293 , Células HT29 , Humanos , Ratones Desnudos , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/genética , Péptidos/metabolismo , Interferencia de ARN , Transducción de Señal , Factores de Tiempo , Transfección , Carga Tumoral , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
4.
J Pathol ; 225(2): 305-14, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21826669

RESUMEN

Several in vitro assays have been proposed to identify cancer stem cells (CSCs), including immunophenotyping, sphere assay and side population (SP) assay. CD133 antigen has been proposed as a CSC marker in colon cancer (CC). However, no functional data are available to date and conflicting results have been reported regarding its role as true CSC marker. Here we set out to identify a molecular signature associated with potential CSC. CD133(+) cells isolated from the CaCo-2 CC cell line were analysed by microarray molecular profiling compared to CD133(-) counterparts. Various differentially expressed genes were identified and the most relevant transcripts found to be over-expressed in CD133(+) cells were evaluated by quantitative RT-PCR in the CD133(+) fractions isolated from several CC cell lines. In the attempt to find a correlation between putative CSCs, isolated by means of CD133 immunophenotyping and the SP approach, we demonstrated a significant enrichment of CD133(+) cells within the SP fraction of CC cells, and comparison of the gene expression profiles revealed that Endothelin-1 (END-1) and nuclear receptor subfamily 4, group A, member 2 (NR4A2) transcripts are highly expressed in both CD133(+) and SP fractions of CC cells. Moreover, depletion of CD133 by siRNA induced a significant attenuation of END-1 and NR4A2 expression levels in CaCo-2 cells, while expression of all three molecules decreased during sodium butyrate-induced differentiation. In conclusion, we have identified a molecular signature associated with potential CSCs and showed for the first time the existence of a functional relationship between CD133, END-1 and NR4A2 expression in colon cancer cells.


Asunto(s)
Antígenos CD/genética , Neoplasias del Colon/genética , Endotelina-1/genética , Regulación Neoplásica de la Expresión Génica/genética , Glicoproteínas/genética , Células Madre Neoplásicas/metabolismo , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Péptidos/genética , Antígeno AC133 , Antígenos CD/metabolismo , Western Blotting , Células CACO-2 , Separación Celular , Neoplasias del Colon/metabolismo , Endotelina-1/metabolismo , Citometría de Flujo , Perfilación de la Expresión Génica , Glicoproteínas/metabolismo , Humanos , Inmunofenotipificación , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Péptidos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
J Biomed Biotechnol ; 2011: 468705, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22219658

RESUMEN

Due to its abundance, easy retrieval, and plasticity characteristics, adipose-tissue-derived stromal cells (ATSCs) present unquestionable advantages over other adult-tissue-derived stem cells. Based on the in silico analysis of our previous data reporting the ATSC-specific expression profiles, the present study attempted to clarify and validate at the functional level the expression of the neurospecific genes expressed by ATSC both in vitro and in vivo. This allowed evidencing that ATSCs express neuro-specific trophins, metabolic genes, and neuroprotective molecules. They were in fact able to induce neurite outgrowth in vitro, along with tissue-specific commitment along the neural lineage and the expression of the TRKA neurotrophin receptor in vivo. Our observation adds useful information to recent evidence proposing these cells as a suitable tool for cell-based applications in neuroregenerative medicine.


Asunto(s)
Tejido Adiposo/citología , Moléculas de Adhesión Celular/metabolismo , Técnicas de Cocultivo/métodos , Neuritas/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Células del Estroma/metabolismo , Tejido Adiposo/metabolismo , Adulto , Animales , Diferenciación Celular , Células Cultivadas , Medios de Cultivo Condicionados , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Masculino , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Persona de Mediana Edad , Células PC12 , Cultivo Primario de Células , Ratas , Regulación hacia Arriba
6.
J Biomed Biotechnol ; 2011: 860578, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22228987

RESUMEN

Mesenchymal stem cells (MSCs), represent an attractive tool for the establishment of a successful stem-cell-based therapy of liver diseases. A number of different mechanisms contribute to the therapeutic effects exerted by MSCs, since these cells can differentiate into functional hepatic cells and can also produce a series of growth factors and cytokines able to suppress inflammatory responses, reduce hepatocyte apoptosis, regress liver fibrosis, and enhance hepatocyte functionality. To date, the infusion of MSCs or MSC-conditioned medium has shown encouraging results in the treatment of fulminant hepatic failure and in end-stage liver disease in experimental settings. However, some issues under debate hamper the use of MSCs in clinical trials. This paper summarizes the biological relevance of MSCs and the potential benefits and risks that can result from translating the MSC research to the treatment of liver diseases.


Asunto(s)
Hepatopatías/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Humanos , Sistema Inmunológico/inmunología , Hepatopatías/patología , Trasplante de Hígado , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Células Madre Mesenquimatosas/inmunología , Factores de Riesgo
7.
Oncotarget ; 7(33): 53047-53063, 2016 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-27344175

RESUMEN

Platelet derived growth factor receptors (PDGFRs) play an important role in tumor pathogenesis, and they are frequently overexpressed in glioblastoma (GBM). Earlier we have shown a higher protein expression of PDGFR isoforms (α and ß) in peritumoral-tissue derived cancer stem cells (p-CSC) than in tumor core (c-CSC) of several GBM affected patients. In the current study, in order to assess the activity of PDGFRα/PDGF-AA signaling axis, we performed time course experiments to monitor the effects of exogenous PDGF-AA on the expression of downstream target genes in c-CSC vs p-CSC. Interestingly, in p-CSC we detected the upregulation of Y705-phosphorylated Stat3, concurrent with a decrement of Rb1 protein in its active state, within minutes of PDGF-AA addition. This finding prompted us to elucidate the role of PDGFRα in self-renewal, invasion and differentiation in p-CSC by using short hairpin RNA depletion of PDGFRα expression. Notably, in PDGFRα-depleted cells, protein analysis revealed attenuation of stemness-related and glial markers expression, alongside early activation of the neuronal marker MAP2a/b that correlated with the induction of tumor suppressor Rb1. The in vitro reduction of the invasive capacity of PDGFRα-depleted CSC as compared to parental cells correlated with the downmodulation of markers of epithelial-mesenchymal transition phenotype and angiogenesis. Surprisingly, we observed the induction of anti-apoptotic proteins and compensatory oncogenic signals such as EDN1, EDNRB, PRKCB1, PDGF-C and PDGF-D. To conclude, we hypothesize that the newly discovered PDGFRα/Stat3/Rb1 regulatory axis might represent a potential therapeutic target for GBM treatment.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Células Madre Neoplásicas/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas de Unión a Retinoblastoma/metabolismo , Factor de Transcripción STAT3/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Adulto , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Células Cultivadas , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/genética , Glioblastoma/patología , Humanos , Células Madre Neoplásicas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/farmacología , Interferencia de ARN , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
8.
Sci Rep ; 5: 9149, 2015 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-25779766

RESUMEN

Although the only effective drug against primary hepatocarcinoma, the multikinase inhibitor Sorafenib (SFB) usually fails to eradicate liver cancer. Since SFB targets mitochondria, cell metabolic reprogramming may underlie intrinsic tumor resistance. To characterize cancer cell metabolic response to SFB, we measured oxygen consumption, generation of reactive oxygen species (ROS) and ATP content in rat LCSC (Liver Cancer Stem Cells) -2 cells exposed to the drug. Genome wide analysis of gene expression was performed by Affymetrix technology. SFB cytotoxicity was evaluated by multiple assays in the presence or absence of metabolic inhibitors, or in cells genetically depleted of mitochondria. We found that low concentrations (2.5-5 µM) of SFB had a relatively modest effect on LCSC-2 or 293 T cell growth, but damaged mitochondria and increased intracellular ROS. Gene expression profiling of SFB-treated cells was consistent with a shift toward aerobic glycolysis and, accordingly, SFB cytotoxicity was dramatically increased by glucose withdrawal or the glycolytic inhibitor 2-DG. Under metabolic stress, activation of the AMP dependent Protein Kinase (AMPK), but not ROS blockade, protected cells from death. We conclude that mitochondrial damage and ROS drive cell killing by SFB, while glycolytic cell reprogramming may represent a resistance strategy potentially targetable by combination therapies.


Asunto(s)
Antineoplásicos/farmacología , Niacinamida/análogos & derivados , Compuestos de Fenilurea/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Autofagia , Línea Celular Tumoral , Respiración de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Desoxiglucosa/farmacología , Metabolismo Energético/efectos de los fármacos , Glucólisis/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Niacinamida/farmacología , Ratas , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Sorafenib , Serina-Treonina Quinasas TOR/metabolismo
9.
Biomed Res Int ; 2014: 859871, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24527460

RESUMEN

There is an emerging body of evidence that chemoresistance and minimal residual disease result from selective resistance of a cell subpopulation from the original tumor that is molecularly and phenotypically distinct. These cells are called "cancer stem cells" (CSCs). In this review, we analyze the potential targeting strategies for eradicating CSCs specifically in order to develop more effective therapeutic strategies for metastatic colon cancer. These include induction of terminal epithelial differentiation of CSCs or targeting some genes expressed only in CSCs and involved in self-renewal and chemoresistance. Ideal targets could be cell regulators that simultaneously control the stemness and the resistance of CSCs. Another important aspect of cancer biology, which can also be harnessed to create novel broad-spectrum anticancer agents, is the Warburg effect, also known as aerobic glycolysis. Actually, little is yet known with regard to the metabolism of CSCs population, leaving an exciting unstudied avenue in the dawn of the emerging field of metabolomics.


Asunto(s)
Neoplasias del Colon , Sistemas de Liberación de Medicamentos , Resistencia a Antineoplásicos , Células Madre Neoplásicas , Animales , Antineoplásicos , Línea Celular Tumoral , Humanos , Ratones
10.
World J Gastroenterol ; 19(29): 4671-8, 2013 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-23922464

RESUMEN

Among somatic stem cells, those residing in the intestine represent a fascinating and poorly explored research field. Particularly, somatic stem cells reside in the small intestine at the level of the crypt base, in a constant balance between self-renewal and differentiation. Aim of the present review is to delve into the mechanisms that regulate the delicate equilibrium through which intestinal stem cells orchestrate intestinal architecture. To this aim, special focus will be addressed to identify the integrating signals from the surrounding niche, supporting a model whereby distinct cell populations facilitate homeostatic vs injury-induced regeneration.


Asunto(s)
Enfermedades Intestinales , Mucosa Intestinal , Intestinos , Células Madre , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Humanos , Enfermedades Intestinales/metabolismo , Enfermedades Intestinales/patología , Enfermedades Intestinales/fisiopatología , Mucosa Intestinal/metabolismo , Intestinos/patología , Intestinos/fisiopatología , Regeneración , Transducción de Señal , Nicho de Células Madre , Células Madre/metabolismo , Células Madre/patología
11.
World J Gastroenterol ; 19(20): 2997-3006, 2013 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-23716979

RESUMEN

Tumors have long been viewed as a population in which all cells have the equal propensity to form new tumors, the so called conventional stochastic model. The cutting-edge theory on tumor origin and progression, tends to consider cancer as a stem cell disease. Stem cells are actively involved in the onset and maintenance of colon cancer. This review is intended to examine the state of the art on colon cancer stem cells (CSCs), with regard to the recent achievements of basic research and to the corresponding translational consequences. Specific prominence is given to the hypothesized origin of CSCs and to the methods for their identification. The growing understanding of CSC biology is driving the optimization of novel anti-cancer targeted drugs.


Asunto(s)
Neoplasias del Colon/patología , Células Madre Neoplásicas/patología , Animales , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/metabolismo , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/metabolismo , Humanos , Terapia Molecular Dirigida , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Transducción de Señal
13.
Dig Liver Dis ; 42(12): 895-901, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20605541

RESUMEN

BACKGROUND: Adipose tissue-derived stromal cells (ATSCs) hold great promises in regenerative medicine. In the last decade, several studies have reported the plasticity of ATSCs toward a hepatocyte-like phenotype. Nonetheless, the molecular mechanisms underlying the conversion from a mesenchymal to an epithelial phenotype remain poorly understood. AIM: In this study, we compared the full genome expression profiles of ATSCs cultured for 4 weeks under pro-hepatogenic conditions to undifferentiated ATSCs, in order to depict the molecular events involved in ATSC hepatic transdifferentiation. METHODS: Analysis was performed using the Affymetrix human focus arrays. Sets of differentially expressed genes were functionally categorized in order to understand which pathways drive the hepatic conversion and interesting targets were validated by Q-PCR. RESULTS: ATSC-derived hepatocyte-like cells activate several genes associated with specific liver functions, including protein metabolism, innate immune response regulation, and biodegradation of toxic compounds. Furthermore, microarray analysis highlighted downregulation of transcripts associated with the mesenchymal lineage, while epithelial-related genes were overexpressed. CONCLUSION: Our data suggest that the in vitro system used in this study drove ATSCs toward a hepatic conversion through a subtle regulation of molecular pathways controlling lineage commitment that promote mesenchymal-epithelial transition.


Asunto(s)
Tejido Adiposo/citología , Hepatocitos/citología , Células del Estroma/citología , Diferenciación Celular/genética , Hepatocitos/fisiología , Humanos , Fenotipo , Células del Estroma/fisiología
14.
J Endocrinol ; 198(3): 471-87, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18550786

RESUMEN

There is evidence that tissue-specific stem cells reside in certain adult tissues. Their specific properties remain elusive, because they are rare and heterogeneous in parent tissues; furthermore, technical difficulties have been encountered in the identification and characterization of their progeny. The aim of this study was to isolate stem/progenitor cells from the human thyroid. We devised a method based on the enzymatic digestion of fresh surgical thyroid specimens, followed by culture of cells in the presence of epidermal growth factor and basic fibroblast growth factor. We also used markers that identify and characterize these cells. Spheroids with self-replicative potential were obtained from all thyroid specimens. The isolated population contained a subset of CD34+ CD45- cells and it was able, in differentiation conditions, to generate follicles with thyroid hormonal production. In support of the plasticity concept, we obtained evidence that, when most freshly isolated spheroids were co-cultured with a neuroblastoma cell line, they produced progeny expressing the neuronal marker beta-tubulin III. Spheroids were also able to undergo adipogenic differentiation when cultured in adipogenic medium. We conclude that a predominant functional type of stem/progenitor cell exists within the thyroid, with an intrinsic ability to generate thyroidal cells and the potential to produce non-thyroidal cells.


Asunto(s)
Esferoides Celulares/citología , Células Madre/citología , Glándula Tiroides/citología , Antígenos CD34/inmunología , Técnicas de Cultivo de Célula , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Factor de Crecimiento Epidérmico/farmacología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Citometría de Flujo , Humanos , Inmunohistoquímica , Inmunofenotipificación , Antígenos Comunes de Leucocito/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/inmunología , Esferoides Celulares/metabolismo , Células Madre/efectos de los fármacos , Células Madre/inmunología , Células Madre/metabolismo , Telomerasa/metabolismo , Telómero/genética
15.
J Autoimmun ; 26(3): 197-207, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16549322

RESUMEN

By screening random peptide libraries (RPLs) with sera of Type 1 diabetes (T1D) patients, we previously identified 5 disease-specific 'mimotopes' displayed on phages (phagotopes). We already characterised 1 phagotope (CH1p), as an epitope of human osteopontin, an autoantigen expressed within the somatostatin cells of human islets. In this paper, we report the characterization of the second phagotope, 195Dyn, by immunohistochemistry, Western Blotting and screening of a human islet cDNA library using rabbit anti-195Dyn antibodies. The 195Dyn mimotope was detected in human islets. The screening of a lambdagt11 cDNA library from human islets has identified a clone, which corresponded to human importin beta. ELISA detected autoantibodies against this protein in sera of around 60% of TD1 patients and in 30% of patients affected by other autoimmune diseases. In summary, RPLs technology proved again successful in identifying another novel autoantigen (importin beta), whose significance in the autoimmune process remains to be fully elucidated.


Asunto(s)
Autoantígenos/inmunología , beta Carioferinas/inmunología , Animales , Autoantígenos/genética , Secuencia de Bases , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Ensayo de Inmunoadsorción Enzimática/métodos , Humanos , Inmunohistoquímica , Islotes Pancreáticos/citología , Islotes Pancreáticos/inmunología , Datos de Secuencia Molecular , Biblioteca de Péptidos , Conejos , Radioinmunoensayo , beta Carioferinas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA