Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Med Res Rev ; 41(1): 464-506, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33038031

RESUMEN

The vascular endothelial growth factor (VEGF) family and its receptors play fundamental roles not only in physiological but also in pathological angiogenesis, characteristic of cancer progression. Aiming at finding putative treatments for several malignancies, various small molecules, antibodies, or protein-based drugs have been evaluated in vitro and in vivo as VEGF inhibitors, providing efficient agents approved for clinical use. Due to the high clinical importance of VEGF, also a great number of anti-VEGF nucleic acid-based aptamers-that is, oligonucleotides able to bind with high affinity and specificity a selected biological target-have been developed as promising agents in anticancer strategies. Notable research efforts have been made in optimization processes of the identified aptamers, searching for increased target affinity and/or bioactivity by exploring structural analogues of the lead compounds. This review is focused on recent studies devoted to the development of DNA-based aptamers designed to target VEGF. Their therapeutic potential as well as their significance in the construction of highly selective biosensors is here discussed.


Asunto(s)
Aptámeros de Nucleótidos , Neoplasias , ADN , Humanos , Neoplasias/diagnóstico , Neoplasias/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
2.
Am J Med Genet A ; 182(1): 176-182, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31609081

RESUMEN

Dominant Optic Atrophy and Deafness (DOAD) may be associated with one or more of the following disorders such as myopathy, progressive external ophthalmoplegia, peripheral neuropathy, and cerebellar atrophy ("DOA-plus"). Intra- and interfamilial variability of the "DOA-plus" phenotype is frequently observed in the majority of the patients carrying the same mutation in the OPA1 gene. We are describing two familial cases of "DOA-plus" carrying the same c.1334G>A (p.Arg445His) mutation in OPA1 and disclosing different clinical, pathological and biochemical features. The two patients showed different expression levels of the mitochondrial OMI/HTRA2 molecule, which acts as a mitochondrial stress sensor and has been described to interplay with OPA1 in in vitro studies. Our data offer the cue to inquire the role of OMI/HTRA2 as a modifier gene in determining the "DOAplus" phenotype variability.


Asunto(s)
Sordera/genética , GTP Fosfohidrolasas/genética , Serina Peptidasa A2 que Requiere Temperaturas Altas/genética , Atrofia Óptica Autosómica Dominante/genética , Adulto , Sordera/fisiopatología , Femenino , Regulación de la Expresión Génica/genética , Predisposición Genética a la Enfermedad , Humanos , Persona de Mediana Edad , Mitocondrias/genética , Mitocondrias/metabolismo , Enfermedades Musculares/genética , Enfermedades Musculares/fisiopatología , Mutación/genética , Oftalmoplejía Externa Progresiva Crónica/genética , Oftalmoplejía Externa Progresiva Crónica/fisiopatología , Atrofia Óptica Autosómica Dominante/fisiopatología , Linaje , Enfermedades del Sistema Nervioso Periférico/genética , Enfermedades del Sistema Nervioso Periférico/fisiopatología
3.
J Cell Physiol ; 233(9): 6508-6517, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29574790

RESUMEN

Nowadays, epigenetics covers a crucial role in different fields of science. The enhancer of zeste homolog 2 (EZH2), the catalytic subunit of the Polycomb Repressive Complex 2 (PRC2), is a big proponent of how epigenetic changes can affect the initiation and progression of several diseases. Through its catalytic activity, responsible for the tri-methylation of lysine 27 of the histone H3 (H3K27me3), EZH2 is a good target for both diagnosis and therapy of different pathologies. A large number of studies have demonstrated its crucial role in cancer initiation and progression. Nevertheless, only recently its function in virus diseases has been uncovered; therefore, EZH2 can be an important promoter of viral carcinogenesis. This review explores the role of EZH2 in viral epigenetics based on recent progress that demonstrated the role of this protein in virus environment. In particular, the review focuses on EZH2 behavior in Hepatitis B Virus, analyzing its role in the rise of Hepatocellular Carcinoma.


Asunto(s)
Carcinogénesis/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/virología , Proteína Potenciadora del Homólogo Zeste 2/genética , Epigénesis Genética/genética , Virus de la Hepatitis B/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/virología , Animales , Carcinogénesis/patología , Carcinoma Hepatocelular/patología , Hepatitis B/genética , Hepatitis B/virología , Humanos , Neoplasias Hepáticas/patología
4.
J Cell Physiol ; 233(5): 3955-3967, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-28884813

RESUMEN

Aging is a primary risk factor for both neurodegenerative disorders (NDs) and tumors such as adult-onset brain tumors. Since NDs and tumors are severe, disabling, progressive and often incurable conditions, they represent a pressing problem in terms of human suffering and economic costs to the healthcare systems. The current challenge for physicians and researchers is to develop new therapeutic strategies in both areas to improve the patients' quality of life. In addition to genetics and environmental stressors, the increase in cellular oxidative stress as one of the potential common etiologies has been reported for both disorders. Recently, the scientific community has focused on the beneficial effects of dietary antioxidant classes, known as nutraceuticals, such as carotenoids, vitamins, and polyphenols. Among these compounds, polyphenols are considered to be one of the most bioactive agents in neurodegeneration and tumor prevention. Despite the beneficial activity of polyphenols, their poor bioavailability and inefficient delivery systems are the main factors limiting their use in medicine and functional food. The development of polymeric nanoparticle-based delivery systems able to encapsulate and preserve polyphenolic compounds may represent a promising tool to enhance their stability, solubility, and cell membrane permeation. In the present review we provide an overview of the main polyphenolic compounds used for ND and brain tumor prevention and treatment that explores their mechanisms of action, recent clinical findings and principal factors limiting their application in medicine.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Enfermedades Neurodegenerativas/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Polifenoles/uso terapéutico , Antioxidantes/química , Antioxidantes/uso terapéutico , Disponibilidad Biológica , Neoplasias Encefálicas/patología , Permeabilidad de la Membrana Celular/efectos de los fármacos , Humanos , Nanopartículas/química , Nanopartículas/uso terapéutico , Enfermedades Neurodegenerativas/patología , Polifenoles/química
5.
J Cell Physiol ; 233(11): 8996-9006, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29904927

RESUMEN

Mesenchymal stromal cells (MSCs) are considered to be an excellent source in regenerative medicine. They contain several cell subtypes, including multipotent stem cells. MSCs are of particular interest as they are currently being tested using cell and gene therapies for a number of human diseases. They represent a rare population in tissues; for this reason, they require, before being transplanted, an in vitro amplification. This process may induce replicative senescence, thus affecting differentiation and proliferative capacities. Increasing evidence suggests that MSCs from fetal tissues are significantly more plastic and grow faster than MSCs from bone marrow. Here, we compare amniotic fluid mesenchymal stromal cells (AF-MSCs) and bone marrow mesenchymal stromal cells (BM-MSCs) in terms of cell proliferation, surface markers, multidifferentiation potential, senescence, and DNA repair capacity. Our study shows that AF-MSCs are less prone to senescence with respect to BM-MSCs. Moreover, both cell models activate the same repair system after DNA damage, but AF-MSCs are able to return to the basal condition more efficiently with respect to BM-MSCs. Indeed, AF-MSCs are better able to cope with genotoxic stress that may occur either during in vitro cultivation or following transplantation in patients. Our findings suggest that AF-MSCs may represent a valid alternative to BM-MSCs in regenerative medicine, and, of great relevance, the investigation of the mechanisms involved in DNA repair capacity of both AF-MSCs and BM-MSCs may pave the way to their rational use in the medical field.


Asunto(s)
Líquido Amniótico/metabolismo , Proliferación Celular/genética , Senescencia Celular/genética , Células Madre Mesenquimatosas/citología , Líquido Amniótico/citología , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular/genética , Células Cultivadas , Femenino , Humanos , Células Madre Mesenquimatosas/metabolismo , Células Madre Multipotentes/citología , Células Madre Multipotentes/metabolismo
6.
J Cell Physiol ; 233(8): 5829-5837, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29215735

RESUMEN

Autosomal recessive Pompe disease is a lysosomal disorder caused by mutations of the acid-α-glucosidase (GAA) gene. Deficiency of GAA enzyme leads to glycogen accumulation and autophagy impairment in cardiac and skeletal muscles, but also in lymphocytes. Since an effective therapy is available, a rapid, sensitive, and specific test is crucial to early identify affected subjects. Number of lymphocytes containing PAS-positive vacuoles was evaluated on blood films from 72 consecutive adult patients with hyperckemia and/or muscle weakness, 13 genetically confirmed late-onset-Pompe-disease (LOPD) and 13 of their offspring. GAA activity, measured on dried blood spot (DBS) in all patients inversely correlated with number of PAS-positive lymphocytes. More than 4 PAS-positive lymphocytes were found in 11 out of the 72 patients (6 new diagnosis of LOPD, 3 different glycogen storage myopathies, 1 glucose-6-phosphate dehydrogenase deficiency, 1 caveolinopathy), in all 13 LOPD patients and in the 13 LOPD offspring. These latter resulted to have all a single GAA mutation but low GAA levels. Immunostaining with the autophagy markers LC3 and p62 confirmed the autophagic nature of lymphocytes vacuoles. ROC curve assessment of PAS-positive lymphocytes disclosed 100% of sensitivity and 94% of specificity in recognizing both compound heterozygous and heterozygous GAA carriers. The other myopathies with more than 4 PAS-positive lymphocytes appeared to be all related to impaired autophagy, which seems to be responsible of PAS-positive vacuolated lymphocytes formation. Quantification of PAS-positive lymphocytes in blood films is useful to identify autophagic vacuolar myopathies and should be routinely used as first level test for Pompe disease.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II/diagnóstico , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Linfocitos/metabolismo , Vacuolas/patología , alfa-Glucosidasas/genética , Adolescente , Adulto , Anciano , Autofagia/fisiología , Niño , Femenino , Humanos , Lisosomas/patología , Masculino , Persona de Mediana Edad , Músculo Esquelético/patología , Adulto Joven
7.
Neurol Sci ; 39(2): 275-285, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29101592

RESUMEN

Neuroglobin (Ngb) is expressed in the central and peripheral nervous system, cerebrospinal fluid, retina, and endocrine tissues where it is involved in binding O2 and other gasotransmitters. Several studies have highlighted its endogenous neuroprotective function. Huntington's disease (HD), a dominant hereditary disease, is characterized by the gradual loss of neurons in discrete areas of the central nervous system. We analyzed the expression of Ngb in the brain tissue of a mouse model of HD, in order to define the role of Ngb with respect to individual cell type vulnerability in HD and to gender and age of mice. Our results showed different expressions of Ngb among neurons of a specific region and between different brain regions. We evidenced a decreased intensity of Ngb at 13 weeks of age, compared to 7 weeks of age. The double immunofluorescence and fluorescence resonance energy transfer (FRET) experiments showed that the co-localization between Ngb and huntingtin at the subcellular level was not close enough to account for a direct interaction. We also observed a different expression of Ngb in the striatum, depending on the sex and age of animals. These findings provide the first experimental evidence for an adaptive response of Ngb in HD, suggesting that Ngb may exert neuroprotective effects in HD beyond its role in reducing sensitivity to oxidative stress.


Asunto(s)
Cuerpo Estriado/metabolismo , Regulación de la Expresión Génica/genética , Globinas/metabolismo , Enfermedad de Huntington/patología , Proteínas del Tejido Nervioso/metabolismo , Factores de Ribosilacion-ADP , Animales , Toxinas Bacterianas , Línea Celular Tumoral , Colinesterasas/metabolismo , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Femenino , Transferencia Resonante de Energía de Fluorescencia , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Masculino , Ratones , Ratones Transgénicos , Mutación/genética , Neuroglobina , Neuronas/metabolismo , Parvalbúminas/metabolismo , Factores Sexuales , Factores de Tiempo
8.
Med Res Rev ; 36(5): 845-70, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27114232

RESUMEN

In familial neurodegenerative disorders, protein aggregates form continuously because of genetic mutations that drive the synthesis of truncated or unfolded proteins. The oxidative stress imposed by neurotransmitters and environmental neurotoxins constitutes an additional threat to the folding of the proteins and the integrity of organelle membranes in neurons. Failure in degrading such altered materials compromises the function of neurons and eventually leads to neurodegeneration. The lysosomal proteolytic enzyme Cathepsin D is the only aspartic-type protease ubiquitously expressed in all the cells of the human body, and it is expressed at high level in the brain. In general, cathepsin D mediated proteolysis is essential to neuronal cell homeostasis through the degradation of unfolded or oxidized protein aggregates delivered to lysosomes via autophagy or endocytosis. More specifically, many altered neuronal proteins that hallmark neurodegenerative diseases (e.g., the amyloid precursor, α-synuclein, and huntingtin) are physiologic substrates of cathepsin D and would abnormally accumulate if not efficiently degraded by this enzyme. Furthermore, experimental evidence indicates that cathepsin D activity is linked to the metabolism of cholesterol and of glycosaminoglycans, which accounts for its involvement in neuronal plasticity. This review focuses on the unique role of cathepsin D mediated proteolysis in the pathogenesis of human neurodegenerative diseases.


Asunto(s)
Catepsina D/metabolismo , Enfermedades Neurodegenerativas/enzimología , Animales , Humanos , Enfermedades Neurodegenerativas/etiología
9.
J Neurochem ; 135(6): 1123-8, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26478990

RESUMEN

Neurofibromatosis type 1 (NF1) is an autosomal dominant genetic condition caused by dominant loss-of-function mutations of the tumor suppressor gene NF1 that encodes neurofibromin, a negative regulator of RAS activity. Mutation analysis of NF1 located at 17q11.2 has been hampered by the large size of the gene, the high rate of new mutations, the lack of mutational clustering, and the presence of several homologous loci. To date, about 80% of the reported NF1 mutations are predicted to result in protein truncation, but very few studies have correlated the causative NF1 mutation with its effect at the protein level. We evaluated a novel diagnostic method to detect truncated forms of neurofibromin in a large cohort of unrelated subjects suspected of having NF1, according to the NIH consensus criteria. Western blot analysis was carried out on protein extracts from patients' leukocytes to highlight the possible presence of altered neurofibromin as a result of mutations in NF1. Truncated neurofibromin was identified in 274/336 patients (81%), confirming the usefulness and reproducibility of the proposed diagnostic approach. Our methodology can be routinely applied in the diagnostic setting, thanks to its simplicity and reliability. Combined with molecular approaches, it may increase the accuracy and efficiency of NF1 genetic testing. We evaluated a novel diagnostic method to detect truncated forms of neurofibromin in patients fulfilling the clinical criteria for Neurofibromatosis 1. Western blot analysis identified truncated neurofibromin in 274/336 patients (81%). Our results indicate that the proposed technique is cheap and reliable, and could ideally be performed as a preliminary biochemical screening before molecular analysis of the NF1 gene.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Mutación/genética , Neurofibromatosis 1/diagnóstico , Neurofibromatosis 1/metabolismo , Neurofibromina 1/genética , Neurofibromina 1/metabolismo , Adolescente , Adulto , Anciano , Niño , Análisis Mutacional de ADN , Femenino , Genes de Neurofibromatosis 1/fisiología , Pruebas Genéticas/métodos , Humanos , Masculino , Persona de Mediana Edad , Reproducibilidad de los Resultados , Adulto Joven
10.
Biochim Biophys Acta ; 1832(1): 105-13, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22974559

RESUMEN

Huntingtin (htt) is a scaffold protein localized at the subcellular level and is involved in coordinating the activity of several protein for signaling and intracellular transport. The emerging properties of htt in intracellular trafficking prompted us to study the role of mutant htt (polyQ-htt) in the intracellular fate of epidermal growth factor receptor (EGFR), whose activity seems to be strictly regulated by htt. In particular, to evaluate whether protein trafficking dysfunction occurs in non-neuronal cells in the absence of functional htt, we monitored the EGFR protein in fibroblasts from homozygotic HD patients and their healthy counterpart. We found that polyQ-htt controls EGFR degradation and recycling. Lack of wild-type htt caused alteration of the ubiquitination cycle, formation of EGFR-incorporating high-molecular weight protein aggregates and abnormal EGFR distribution in endosomes of the degradation and recycling pathways after EGF stimulation. PolyQ-htt-induced alteration of EGFR trafficking affected cell migration and proliferation, at least in part, through inhibition of ERK signaling. To our knowledge the data here reported represent the first signaling and phenotypic characterization of polyQ-htt involvement in the modulation of growth factor stimulation in non-neuronal cells.


Asunto(s)
Receptores ErbB/metabolismo , Fibroblastos/metabolismo , Enfermedad de Huntington/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Adulto , Células Cultivadas , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/genética , Femenino , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Masculino , Persona de Mediana Edad , Mutación , Fosforilación , Transporte de Proteínas
11.
Neurobiol Dis ; 52: 104-16, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23220622

RESUMEN

In Huntington's disease (HD) mutant huntingtin protein impairs the function of several transcription factors, in particular the cAMP response element-binding protein (CREB). CREB activation can be increased by targeting phosphodiesterases such as phospohodiesterase 4 (PDE4) and phosphodiesterase 10A (PDE10A). Indeed, both PDE4 inhibition (DeMarch et al., 2008) and PDE10A inhibition (Giampà et al., 2010) proved beneficial in the R6/2 mouse model of HD. However, Hebb et al. (2004) reported PDE10A decline in R6/2 mice. These findings raise the issue of how PDE10A inhibition is beneficial in HD if such enzyme is lost. R6/2 mice and their wild type littermates were treated with the PDE10A inhibitor TP10 (a gift from Pfizer) or saline, sacrificed at 5, 9, and 13 weeks of age, and single and double label immunohistochemistry and western blotting were performed. PDE10A increased dramatically in the spiny neurons of R6/2 compared to the wild type mice. Conversely, in the striatal cholinergic interneurons, PDE10A was lower and it did not change significantly with disease progression. In the other subsets of striatal interneurons (namely, parvalbuminergic, somatostatinergic, and calretininergic interneurons) PDE10A immunoreactivity was higher in the R6/2 compared to the wild-type mice. In the TP10 treated R6/2, PDE10A levels were lower than in the saline treated mice in the medium spiny neurons, whereas they were higher in all subsets of striatal interneurons except for the cholinergic ones. However, in the whole striatum densitometry studies, PDE10A immunoreactivity was lower in the R6/2 compared to the wild-type mice. Our study demonstrates that PDE10A is increased in the spiny neurons of R6/2 mice striatum. Thus, the accumulation of PDE10A in the striatal projection neurons, by hydrolyzing greater amounts of cyclic nucleotides, is likely to contribute to cell damage in HD. Consequently, the beneficial effect of TP10 in HD models (Giampà et al., 2009, 2010) is explained by the efficiency of such compound in counteracting this phenomenon and therefore increasing the availability of cyclic nucleotides.


Asunto(s)
Cuerpo Estriado/enzimología , Enfermedad de Huntington/enzimología , Neuronas/enzimología , Hidrolasas Diéster Fosfóricas/metabolismo , Animales , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Inhibidores de Fosfodiesterasa/farmacología , Hidrolasas Diéster Fosfóricas/genética , Pirazoles/farmacología , Quinolinas/farmacología
12.
Eur J Hum Genet ; 31(8): 931-938, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37217626

RESUMEN

Spinal neurofibromatosis (SNF) is a form of neurofibromatosis type 1 (NF1) characterized by bilateral neurofibromas involving all spinal roots. The pathogenic mechanisms determining the SNF form are currently unknown. To verify the presence of genetic variants possibly related to SNF or classic NF1, we studied 106 sporadic NF1 and 75 SNF patients using an NGS panel of 286 genes encoding RAS pathway effectors and neurofibromin interactors and evaluated the expression of syndecans (SDC1, SDC2, SDC3, SDC4), the NF1 3' tertile interactors, by quantitative real-time PCR. We previously identified 75 and 106 NF1 variants in SNF and NF1 cohorts, respectively. The analysis of the distribution of pathogenic NF1 variants in the three NF1 tertiles showed a significantly higher prevalence of NF1 3' tertile mutations in SNF than in the NF1 cohort. We hypothesized a potential pathogenic significance of the 3' tertile NF1 variants in SNF. The analysis of syndecan expression on PBMCs RNAs from 16 SNF, 16 classic NF1 patients and 16 healthy controls showed that the expression levels of SDC2 and SDC3 were higher in SNF and NF1 patients than in controls; moreover, SDC2, SDC3 and SDC4 were significantly over expressed in patients mutated in the 3' tertile compared to controls. Two different mutational NF1 spectra seem to characterize SNF and classic NF1, suggesting a pathogenic role of NF1 3' tertile and its interactors, syndecans, in SNF. Our study, providing new insights on a possible role of neurofibromin C-terminal in SNF, could address effective personalized patient management and treatments.


Asunto(s)
Neurofibromatosis , Neurofibromatosis 1 , Humanos , Neurofibromatosis 1/genética , Neurofibromina 1/genética , Mutación , Sindecanos/genética , Genes de Neurofibromatosis 1
13.
Neurobiol Dis ; 46(1): 225-33, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22311347

RESUMEN

The mitogen-activated protein kinases (MAPKs) superfamily comprises three major signaling pathways: the extracellular signal-regulated protein kinases (ERKs), the c-Jun N-terminal kinases or stress-activated protein kinases (JNKs/SAPKs) and the p38 family of kinases. ERK 1/2 signaling has been implicated in a number of neurodegenerative disorders, including Huntington's disease (HD). Phosphorylation patterns of ERK 1/2 and JNK are altered in cell models of HD. In this study, we aimed at studying the correlations between ERK 1/2 and the neuronal vulnerability to HD degeneration in the R6/2 transgenic mouse model of HD. Single and double-label immunofluorescence for phospho-ERK (pERK, the activated form of ERK) and for each of the striatal neuronal markers were employed on perfusion-fixed brain sections from R6/2 and wild-type mice. Moreover, Phosphodiesterase 4 inhibition through rolipram was used to study the effects on pERK expression in the different types of striatal neurons. We completed our study with western blot analysis. Our study shows that pERK levels increase with age in the medium spiny striatal neurons and in the parvalbumin interneurons, and that rolipram counteracts such increase in pERK. Conversely, cholinergic and somatostatinergic interneurons of the striatum contain higher levels of pERK in the R6/2 mice compared to the controls. Rolipram induces an increase in pERK expression in these interneurons. Thus, our study confirms and extends the concept that the expression of phosphorylated ERK 1/2 is related to neuronal vulnerability and is implicated in the pathophysiology of cell death in HD.


Asunto(s)
Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/enzimología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Inhibidores de Fosfodiesterasa 4/farmacología , Rolipram/farmacología , Animales , Modelos Animales de Enfermedad , Enfermedad de Huntington/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos
14.
J Cell Sci ; 123(Pt 24): 4332-9, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21098642

RESUMEN

During peripheral nerve injury, Schwann cells (SCs) adopt a migratory phenotype and remodel the extracellular matrix and provide a supportive activity for neuron regeneration. SCs synthesize neurotrophic factors and cytokines that are crucial for the repair of the injured nerve. The receptor for advanced glycation end products (RAGE) and its ligand S100B, which are secreted by SCs, are required for the repair of the injured peripheral nerve in vivo. However, the precise intracellular pathways involved have not been completely elucidated. Here, we show that RAGE-induced S100B secretion involves the recruitment of S100B in lipid rafts and caveolae. Moreover, we demonstrate for the first time that RAGE induces the expression of thioredoxin interacting protein (TXNIP) in SCs and the injured sciatic nerve in vivo. TXNIP is involved in the activation of p38 MAPK, CREB and NFκB in SCs. TXNIP silencing partially inhibits RAGE-induced SC migration and completely abolishes RAGE-induced fibronectin and IL-1ß expression. Our results support a model in which TXNIP mediates in part RAGE-induced SC migration and is required for the expression of provisional ECM and pro-inflammatory IL-1ß. We provide new insight on the role of the SC RAGE-TXNIP axis in the repair of injured peripheral nerves.


Asunto(s)
Proteínas Portadoras/metabolismo , Movimiento Celular , Fibronectinas/metabolismo , Interleucina-1beta/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Receptores Inmunológicos/metabolismo , Proteínas S100/metabolismo , Células de Schwann/citología , Animales , Proteínas de Ciclo Celular , Activación Enzimática , Masculino , Microdominios de Membrana/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor para Productos Finales de Glicación Avanzada , Subunidad beta de la Proteína de Unión al Calcio S100 , Células de Schwann/enzimología , Nervio Ciático/metabolismo , Nervio Ciático/patología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
15.
J Cell Biochem ; 110(4): 903-9, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20564189

RESUMEN

Growth factors and other regulatory molecules are required to direct differentiation of bone marrow-derived human mesenchymal stem cells (hMSC) along specific lineages. However, the therapeutic use of growth factors is limited by their susceptibility to degradation, and the need to maintain prolonged local release of growth factor at levels sufficient to stimulate hMSC. The aim of this study was to investigate whether a device containing heparan sulfate (HS), which is a co-factor in growth factor-mediated cell proliferation and differentiation, could potentiate and prolong the delivery of fibroblast growth factor-2 (FGF-2) and thus enhance hMSC stimulation. To this aim, we synthesized cationic polyelectrolyte polymers covalently and non-covalently anchored to HS and evaluated their effect on hMSC proliferation. Polymers non-covalently bound to HS resulted in the release of an HS/FGF-2 complex rather than FGF-2 alone. The release of this complex significantly restored hMSC proliferation, which was abolished in serum-free medium and only partially restored by the release of FGF-2 alone as occurred with polymer covalently bound to HS. We also demonstrate that exposure to HS/FGF-2 during early growth but not during post-confluence is essential for hMSC differentiation down the fibroblast lineage, which suggests that both factors are required to establish the correct stem cell commitment that is necessary to support subsequent differentiation. In conclusion, the delivery platform described here is a step towards the development of a new class of biomaterial that enables the prolonged, non-covalent binding and controlled delivery of growth factors and cofactors without altering their potency.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Electrólitos/química , Factor 2 de Crecimiento de Fibroblastos/administración & dosificación , Heparitina Sulfato/administración & dosificación , Secuencia de Bases , Cationes , Linaje de la Célula , Células Cultivadas , Cartilla de ADN , Factor 2 de Crecimiento de Fibroblastos/farmacocinética , Factor 2 de Crecimiento de Fibroblastos/farmacología , Heparitina Sulfato/farmacocinética , Heparitina Sulfato/farmacología , Humanos , Células Madre Mesenquimatosas , Polímeros , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Elife ; 92020 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-32223893

RESUMEN

Senescent cells secrete several molecules, collectively named senescence-associated secretory phenotype (SASP). In the SASP of cells that became senescent following several in vitro chemical and physical stress, we identified the IGFBP-4 protein that can be considered a general stress mediator. This factor appeared to play a key role in senescence-paracrine signaling. We provided evidences showing that genotoxic injury, such as low dose irradiation, may promote an IGFBP-4 release in bloodstream both in mice irradiated with 100 mGy X-ray and in human subjects that received Computer Tomography. Increased level of circulating IGFBP-4 may be responsible of pro-aging effect. We found a significant increase of senescent cells in the lungs, heart, and kidneys of mice that were intraperitoneally injected with IGFBP-4 twice a week for two months. We then analyzed how genotoxic stressors may promote the release of IGFBP-4 and the molecular pathways associated with the induction of senescence by this protein.


Asunto(s)
Envejecimiento , Senescencia Celular/genética , Daño del ADN , Proteína 4 de Unión a Factor de Crecimiento Similar a la Insulina/sangre , Proteína 4 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Adolescente , Adulto , Anciano , Animales , Proliferación Celular , Células Cultivadas , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Fenotipo , Transducción de Señal , Tomografía Computarizada por Rayos X , Adulto Joven
17.
Hum Mutat ; 30(5): 741-8, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19242930

RESUMEN

Hyperornithinemia-hyperammonemia-homocitrullinuria (HHH) syndrome is an autosomal recessive disorder of the urea cycle. With the exception of the French-Canadian founder effect, no common mutation has been detected in other populations. In this study, we collected 16 additional HHH cases and expanded the spectrum of SLC25A15/ORC1 mutations. Eleven novel mutations were identified including six new missense and one microrearrangement. We also measured the transport properties of the recombinant purified proteins in reconstituted liposomes for four new and two previously reported missense mutations and proved that the transport activities of these mutant forms of ORC1 were reduced as compared with the wild-type protein; residual activity ranged between 4% and 19%. Furthermore, we designed three-dimensional (3D)-modeling of mutant ORC1 proteins. While modeling the changes in silico allowed us to obtain new information on the pathomechanisms underlying HHH syndrome, we found no clear-cut genotype-phenotype correlations. Although patient metabolic alterations responded well to low-protein therapy, predictions concerning the long-term evolution of HHH syndrome remain uncertain. The preference for a hepatic rather than a neurological presentation at onset also continues, largely, to elude us. Neither modifications in oxidative metabolism-related energy, such as those expected in different mtDNA haplogroups, nor sequence variants in SLC25A2/ORC2 seem to be crucial. Other factors, including protein stability and function, and ORC1-ORC2 structural interactions should be further investigated.


Asunto(s)
Sistemas de Transporte de Aminoácidos Básicos/genética , Citrulina/análogos & derivados , Hiperamonemia/genética , Mutación/genética , Ornitina/sangre , Adulto , Sistemas de Transporte de Aminoácidos Básicos/química , Transporte Biológico , Niño , Preescolar , Citrulina/orina , Escherichia coli , Femenino , Humanos , Hiperamonemia/epidemiología , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Proteínas de Transporte de Membrana Mitocondrial , Proteínas Mutantes/aislamiento & purificación , Estructura Secundaria de Proteína , Síndrome
18.
Neurochem Int ; 117: 174-187, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-28532681

RESUMEN

Parkinsonian-like motor deficits in Huntington's Disease (HD) patients are associated with abnormal dopamine neurotransmission in the striatum. Dopamine metabolism leads to the formation of oxidized dopamine quinones that exacerbates mitochondrial dysfunction with production of reactive oxygen species (ROS) that eventually lead to neuronal cell death. We have previously shown that dopamine-induced oxidative stress triggers apoptotic cell death in dopaminergic neuroblastoma SH-SY5Y cells hyper-expressing the mutant polyQ Huntingtin (polyQ-Htt) protein. Dopamine toxicity was paralleled by impaired autophagy clearance of the polyQ-Htt aggregates. In this study, we found that Dopamine affects the stability and function of ATG4, a redox-sensitive cysteine-protein involved in the processing of LC3, a key step in the formation of autophagosomes. Resveratrol, a dietary polyphenol with anti-oxidant and pro-autophagic properties, has shown neuroprotective potential in HD. Yet the molecular mechanism through which Resveratrol can protect HD cells against DA is not known. Here, we show that Resveratrol prevents the generation of ROS, restores the level of ATG4, allows the lipidation of LC3, facilitates the degradation of polyQ-Htt aggregates and protects the cells from Dopamine toxicity. The present findings provide a mechanistic explanation of the neuroprotective activity of Resveratrol and support its inclusion in a therapeutic regimen to slow down HD progression.


Asunto(s)
Proteínas Relacionadas con la Autofagia/metabolismo , Autofagia/efectos de los fármacos , Cisteína Endopeptidasas/metabolismo , Dopamina/toxicidad , Proteína Huntingtina/biosíntesis , Fármacos Neuroprotectores/farmacología , Fagosomas/efectos de los fármacos , Resveratrol/farmacología , Antioxidantes/farmacología , Autofagia/fisiología , Línea Celular Tumoral , Humanos , Proteína Huntingtina/genética , Mutación/fisiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Fagosomas/metabolismo , Fagosomas/patología
19.
Neuropharmacology ; 135: 126-138, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29526547

RESUMEN

Huntington's disease is a dreadful, incurable disorder. It springs from the autosomal dominant mutation in the first exon of the HTT gene, which encodes for the huntingtin protein (HTT) and results in progressive neurodegeneration. Thus far, all the attempted approaches to tackle the mutant HTT-induced toxicity causing this disease have failed. The mutant protein comes with the aberrantly expanded poly-glutamine tract. It is primarily to blame for the build-up of ß-amyloid-like HTT aggregates, deleterious once broadened beyond the critical ∼35-37 repeats threshold. Recent experimental findings have provided valuable information on the molecular basis underlying this HTT-driven neurodegeneration. These findings indicate that the poly-glutamine siding regions and many post-translation modifications either abet or counter the poly-glutamine tract. This review provides an overall, up-to-date insight into HTT biophysics and structural biology, particularly discussing novel pharmacological options to specifically target the mutated protein and thus inhibit its functions and toxicity.


Asunto(s)
Aptámeros de Nucleótidos/farmacología , Aptámeros de Nucleótidos/uso terapéutico , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/metabolismo , Animales , Humanos , Enfermedad de Huntington/genética , Modelos Moleculares , Procesamiento Proteico-Postraduccional
20.
Exp Mol Med ; 50(3): 1, 2018 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-29563495

RESUMEN

Several aspects of stem cell life are governed by epigenetic variations, such as DNA methylation, histone modifications, and chromatin remodeling. Epigenetic events are also connected with the impairment of stem cell functions. For example, during senescence, there are significant changes in chromatin organization that alter transcription. The MECP2 protein can bind methylated cytosines and contribute to regulating gene expression at one of the highest hierarchical levels. Researchers are particularly interested in this protein, as up to 90% of Rett syndrome patients have an MECP2 gene mutation. Nevertheless, the role of MECP2 in this disease remains poorly understood. We used a mouse model of Rett syndrome to evaluate whether residual MECP2 activity in neural stem cells (NSCs) induced the senescence phenomena that could affect stem cell function. Our study clearly demonstrated that the reduced expression of MECP2 is connected with an increase in senescence, an impairment in proliferation capacity, and an accumulation of unrepaired DNA foci. Mecp2 +/- NSCs did not cope with genotoxic stress in the same way as the control cells did. Indeed, after treatment with different DNA-damaging agents, the NSCs from mice with mutated Mecp2 accumulated more DNA damage foci (γ-H2AX+) and were more prone to cell death than the controls. Senescence in Mecp2 +/- NSCs decreased the number of stem cells and progenitors and gave rise to a high percentage of cells that expressed neither stem/progenitor nor differentiation markers. These cells could be senescent and dysfunctional.


Asunto(s)
Diferenciación Celular/fisiología , Células-Madre Neurales/patología , Síndrome de Rett/patología , Animales , Apoptosis/fisiología , Western Blotting , Ciclo Celular/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Senescencia Celular/fisiología , Daño del ADN/fisiología , Reparación del ADN/fisiología , Modelos Animales de Enfermedad , Femenino , Inmunohistoquímica , Ratones , Células-Madre Neurales/metabolismo , Síndrome de Rett/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA