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










Base de datos
Intervalo de año de publicación
1.
PLoS One ; 18(8): e0275550, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37616231

RESUMEN

BACKGROUND: Renal injury induces major changes in plasma and cardiac metabolites. Using a small- animal in vivo model, we sought to identify a key metabolite whose levels are significantly modified following an acute kidney injury (AKI) and to analyze whether this agent could offer cardiac protection once an ischemic event has occurred. METHODS AND RESULTS: Metabolomics profiling of cardiac lysates and plasma samples derived from rats that underwent AKI 1 or 7 days earlier by 5/6 nephrectomy versus sham-operated controls was performed. We detected 26 differential metabolites in both heart and plasma samples at the two selected time points, relative to sham. Out of which, kynurenic acid (kynurenate, KYNA) seemed most relevant. Interestingly, KYNA given at 10 mM concentration significantly rescued the viability of H9C2 cardiac myoblast cells grown under anoxic conditions and largely increased their mitochondrial content and activity as determined by flow cytometry and cell staining with MitoTracker dyes. Moreover, KYNA diluted in the drinking water of animals induced with an acute myocardial infarction, highly enhanced their cardiac recovery according to echocardiography and histopathology. CONCLUSION: KYNA may represent a key metabolite absorbed by the heart following AKI as part of a compensatory mechanism aiming at preserving the cardiac function. KYNA preserves the in vitro myocyte viability following exposure to anoxia in a mechanism that is mediated, at least in part, by protection of the cardiac mitochondria. A short-term administration of KYNA may be highly beneficial in the treatment of the acute phase of kidney disease in order to attenuate progression to reno-cardiac syndrom and to reduce the ischemic myocardial damage following an ischemic event.


Asunto(s)
Lesión Renal Aguda , Ácido Quinurénico , Animales , Ratas , Ácido Quinurénico/farmacología , Triptófano , Corazón , Hipoxia , Mitocondrias Cardíacas
2.
Vaccines (Basel) ; 10(8)2022 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-35893835

RESUMEN

Patients with inflammatory bowel disease (IBD) treated with anti-tumor-necrosis factor-alpha (TNFα) exhibited lower serologic responses one-month following the second dose of the COVID-19 BNT162b2 vaccine compared to those not treated with anti-TNFα (non-anti-TNFα) or to healthy controls (HCs). We comprehensively analyzed long-term humoral responses, including anti-spike (S) antibodies, serum inhibition, neutralization, cross-reactivity and circulating B cell six months post BNT162b2, in patients with IBD stratified by therapy compared to HCs. Subjects enrolled in a prospective, controlled, multi-center Israeli study received two BNT162b2 doses. Anti-S levels, functional activity, specific B cells, antigen cross-reactivity, anti-nucleocapsid levels, adverse events and IBD disease score were detected longitudinally. In total, 240 subjects, 151 with IBD (94 not treated with anti-TNFα and 57 treated with anti-TNFα) and 89 HCs participated. Six months after vaccination, patients with IBD treated with anti-TNFα had significantly impaired BNT162b2 responses, specifically, more seronegativity, decreased specific circulating B cells and cross-reactivity compared to patients untreated with anti-TNFα. Importantly, all seronegative subjects were patients with IBD; of those, >90% were treated with anti-TNFα. Finally, IBD activity was unaffected by BNT162b2. Altogether these data support the earlier booster dose administration in these patients.

3.
Ocul Surf ; 23: 140-142, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34890804

RESUMEN

Aniridia is a panocular disease causing progressive severe visual impairment and blindness due to PAX-6 haploinsufficiency. One of the most disabling ocular symptoms is aniridia-related keratopathy (ARK), a progressive corneal opacification due to epithelial impairment, vascular and conjunctival pathologies. There is currently no available treatment to prevent progressive visual loss. For this aim, we have used mutant limbal cells for phenotypic screening using FDA-approved and bio-actives drug library and found Duloxetine, a serotonin and norepinephrine reuptake inhibitor used against severe depression as able to enhance endogenous PAX6 expression and target genes, which returned fairly to amounts found in normal limbal cells. In addition, Duloxetine could restore cell migration of the mutant cells. Furthermore, we show that Duloxetine activates PAX6 through inhibition of the ERK pathway on limbal mutant cells. This observation fits the recent report that MEK inhibitors enhance PAX6 in vivo, partially rescuing aniridia developmental phenotype of Pax6+/- mice. The discovery of an unique compound able to enhance PAX6 activity and that could be locally administered using eye drops associated with drug repurposing is expected to lead to rapid development of applicable drugs for the topical (eye drops) treatment of aniridia.


Asunto(s)
Aniridia , Haploinsuficiencia , Animales , Aniridia/genética , Clorhidrato de Duloxetina/farmacología , Proteínas del Ojo/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Soluciones Oftálmicas , Factor de Transcripción PAX6/genética , Transducción de Señal , Células Madre/patología
4.
Biochem Biophys Res Commun ; 582: 100-104, 2021 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-34700241

RESUMEN

Aniridia is a panocular inherited rare eye disease linked to heterozygous mutations on the PAX6 gene, which fail to properly produce sufficient protein essential for normal eye development and function. Most of the patients suffer from aniridia-related keratopathy, a progressive opacification of the cornea. There is no effective treatment for this blinding disease. Here we screen for small compounds and identified Ritanserin, a serotonin 2A receptor antagonist, that can rescue PAX6 haploinsufficiency of mutant limbal cells, defective cell migration and PAX6-target gene expression. We further demonstrated that Ritanserin activates PAX6 production through the selective inactivation of the MEK/ERK signaling pathway. Our data strongly suggest that repurposing this therapeutic molecule could be effective in preventing or treating existing blindness by restoring corneal transparency.


Asunto(s)
Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/genética , Soluciones Oftálmicas/farmacología , Factor de Transcripción PAX6/genética , Ritanserina/farmacología , Antagonistas de la Serotonina/farmacología , Células Madre/efectos de los fármacos , Aniridia/tratamiento farmacológico , Aniridia/genética , Aniridia/metabolismo , Aniridia/patología , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Reposicionamiento de Medicamentos/métodos , Epitelio Corneal/efectos de los fármacos , Epitelio Corneal/metabolismo , Epitelio Corneal/patología , Regulación de la Expresión Génica , Células HEK293 , Haploinsuficiencia , Humanos , Limbo de la Córnea/efectos de los fármacos , Limbo de la Córnea/metabolismo , Limbo de la Córnea/patología , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Modelos Biológicos , Factor de Transcripción PAX6/agonistas , Factor de Transcripción PAX6/metabolismo , Receptor de Serotonina 5-HT2A/genética , Receptor de Serotonina 5-HT2A/metabolismo , Transducción de Señal/efectos de los fármacos , Células Madre/metabolismo , Células Madre/patología
5.
Cells ; 10(6)2021 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-34072893

RESUMEN

Chronic inflammation promotes cancer progression by affecting the tumor cells and their microenvironment. Here, we demonstrate that a continuous stimulation (~6 weeks) of triple-negative breast tumor cells (TNBC) by the proinflammatory cytokines tumor necrosis factor α (TNFα) + interleukin 1ß (IL-1ß) changed the expression of hundreds of genes, skewing the cells towards a proinflammatory phenotype. While not affecting stemness, the continuous TNFα + IL-1ß stimulation has increased tumor cell dispersion and has induced a hybrid metabolic phenotype in TNBC cells; this phenotype was indicated by a transcription-independent elevation in glycolytic activity and by increased mitochondrial respiratory potential (OXPHOS) of TNBC cells, accompanied by elevated transcription of mitochondria-encoded OXPHOS genes and of active mitochondria area. The continuous TNFα + IL-1ß stimulation has promoted in a glycolysis-dependent manner the activation of p65 (NF-kB), and the transcription and protein expression of the prometastatic and proinflammatory mediators sICAM-1, CCL2, CXCL8 and CXCL1. Moreover, when TNBC cells were stimulated continuously by TNFα + IL-1ß in the presence of a glycolysis inhibitor, their conditioned media had reduced ability to recruit monocytes and neutrophils in vivo. Such inflammation-induced metabolic plasticity, which promotes prometastatic cascades in TNBC, may have important clinical implications in treatment of TNBC patients.


Asunto(s)
Medios de Cultivo Condicionados/farmacología , Mediadores de Inflamación/farmacología , Inflamación/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología , Citocinas/genética , Humanos , Inflamación/metabolismo , Mediadores de Inflamación/metabolismo , Fenotipo , Transducción de Señal/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/fisiología , Factor de Necrosis Tumoral alfa/metabolismo
6.
Cancers (Basel) ; 13(6)2021 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-33806906

RESUMEN

The pro-inflammatory cytokines tumor necrosis factor α (TNFα) and interleukin 1ß (IL-1ß) are expressed simultaneously and have tumor-promoting roles in breast cancer. In parallel, mesenchymal stem cells (MSCs) undergo conversion at the tumor site to cancer-associated fibroblasts (CAFs), which are generally connected to enhanced tumor progression. Here, we determined the impact of consistent inflammatory stimulation on stromal cell plasticity. MSCs that were persistently stimulated by TNFα + IL-1ß (generally 14-18 days) gained a CAF-like morphology, accompanied by prominent changes in gene expression, including in stroma/fibroblast-related genes. These CAF-like cells expressed elevated levels of vimentin and fibroblast activation protein (FAP) and demonstrated significantly increased abilities to contract collagen gels. Moreover, they gained the phenotype of inflammatory CAFs, as indicated by the reduced expression of α smooth muscle actin (αSMA), increased proliferation, and elevated expression of inflammatory genes and proteins, primarily inflammatory chemokines. These inflammatory CAFs released factors that enhanced tumor cell dispersion, scattering, and migration; the inflammatory CAF-derived factors elevated cancer cell migration by stimulating the chemokine receptors CCR2, CCR5, and CXCR1/2 and Ras-activating receptors, expressed by the cancer cells. Together, these novel findings demonstrate that chronic inflammation can induce MSC-to-CAF conversion, leading to the generation of tumor-promoting inflammatory CAFs.

7.
Cancer Immunol Immunother ; 69(7): 1337-1352, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32219500

RESUMEN

Cleavage of the MUC1 glycoprotein yields two subunits, an extracellular alpha-subunit bound to a smaller transmembrane beta-subunit. Monoclonal antibodies (mAbs) directed against the MUC1 alpha-beta junction comprising the SEA domain, a stable cell-surface moiety, were generated. Sequencing of all seven anti-SEA domain mAbs showed that they clustered into four groups and sequences of all groups are presented here. mAb DMB5F3 with picomolar affinity for the MUC1 SEA target was selected for further evaluation. Immunohistochemical staining of a series of malignancies with DMB5F3 including lung, prostate, breast, colon, and pancreatic carcinomas revealed qualitative and qualitative differences between MUC1 expression on normal versus malignant cells: DMB5F3 strongly stained malignant cells in a near-circumferential pattern, whereas MUC1 in normal pancreatic and breast tissue showed only weak apical positivity of ductal/acinar cells. Humanized chimeric DMB5F3 linked to ZZ-PE38 (ZZ IgG-binding protein fused to Pseudomonas exotoxin) induced vigorous cytotoxicity of MUC1+ malignant cells in vitro. The intensity of cell killing correlated with the level of MUC1 expression by the target cell, suggesting a MUC1 expression threshold for cell killing. MUC1+ Colo357 pancreatic cancer cells xenotransplanted into nude and SCID mice models were treated with the chDMB5F3:ZZ-PE38 immunocomplex. In both transplant models, chDMB5F3:ZZ-PE38 exhibited significant in vivo anti-tumor activity, suppressing up to 90% of tumor volume in the SCID model compared with concomitant controls. The efficacy of chDMB5F3:ZZ-PE38 immunotoxin in mediating tumor killing both in vitro and in vivo strongly suggests a clinical role for anti-MUC1 SEA antibody in the treatment of MUC1-expressing malignancies.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antineoplásicos Inmunológicos/farmacología , Inmunotoxinas/inmunología , Mucina-1/química , Mucina-1/inmunología , Neoplasias Pancreáticas/tratamiento farmacológico , Animales , Apoptosis , Proliferación Celular , Femenino , Humanos , Ratones , Ratones Desnudos , Ratones SCID , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Dominios Proteicos , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Cell Mol Life Sci ; 77(14): 2795-2813, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31562564

RESUMEN

Neurofibrillary tangles of the Tau protein and plaques of the amyloid ß peptide are hallmarks of Alzheimer's disease (AD), which is characterized by the conversion of monomeric proteins/peptides into misfolded ß-sheet rich fibrils. Halting the fibrillation process and disrupting the existing aggregates are key challenges for AD drug development. Previously, we performed in vitro high-throughput screening for the identification of potent inhibitors of Tau aggregation using a proxy model, a highly aggregation-prone hexapeptide fragment 306VQIVYK311 (termed PHF6) derived from Tau. Here we have characterized a hit molecule from that screen as a modulator of Tau aggregation using in vitro, in silico, and in vivo techniques. This molecule, an anthraquinone derivative named Purpurin, inhibited ~ 50% of PHF6 fibrillization in vitro at equimolar concentration and disassembled pre-formed PHF6 fibrils. In silico studies showed that Purpurin interacted with key residues of PHF6, which are responsible for maintaining its ß-sheets conformation. Isothermal titration calorimetry and surface plasmon resonance experiments with PHF6 and full-length Tau (FL-Tau), respectively, indicated that Purpurin interacted with PHF6 predominantly via hydrophobic contacts and displayed a dose-dependent complexation with FL-Tau. Purpurin was non-toxic when fed to Drosophila and it significantly ameliorated the AD-related neurotoxic symptoms of transgenic flies expressing WT-FL human Tau (hTau) plausibly by inhibiting Tau accumulation and reducing Tau phosphorylation. Purpurin also reduced hTau accumulation in cell culture overexpressing hTau. Importantly, Purpurin efficiently crossed an in vitro human blood-brain barrier model. Our findings suggest that Purpurin could be a potential lead molecule for AD therapeutics.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Antraquinonas/farmacología , Oligopéptidos/genética , Agregado de Proteínas/efectos de los fármacos , Proteínas tau/genética , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/efectos de los fármacos , Animales , Animales Modificados Genéticamente/genética , Barrera Hematoencefálica/efectos de los fármacos , Modelos Animales de Enfermedad , Drosophila melanogaster/genética , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/genética , Fosforilación/efectos de los fármacos , Conformación Proteica en Lámina beta/efectos de los fármacos , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética
9.
Eur J Med Chem ; 179: 779-790, 2019 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-31288127

RESUMEN

Azole antifungals inhibit the biosynthesis of ergosterol, the fungal equivalent of cholesterol in mammalian cells. Here we report an investigation of the activity of coumarin-substituted azole antifungals. Screening against a panel of Candida pathogens, including a mutant lacking CYP51, the target of antifungal azoles, revealed that this enzyme is inhibited by triazole-based antifungals, whereas imidazole-based derivatives have more than one mode of action. The imidazole-bearing antifungals more effectively reduced trailing growth associated with persistence and/or recurrence of fungal infections than triazole-based derivatives. The imidazole derivatives were more toxic to mammalian cells and more potently inhibited the activity of CYP3A4, which is one of the main causes of azole toxicity. Using live cell imaging, we showed that regardless of the type of azole ring fluorescent 7-diethylaminocoumarin-based azoles localized to the endoplasmic reticulum, the organelle that harbors CYP51. This study suggests that the coumarin is a promising scaffold for development of novel azole-based antifungals that effectively localize to the fungal cell endoplasmic reticulum.


Asunto(s)
Antifúngicos/farmacología , Azoles/farmacología , Candida/efectos de los fármacos , Cumarinas/farmacología , Antifúngicos/síntesis química , Antifúngicos/química , Azoles/síntesis química , Azoles/química , Candida/citología , Supervivencia Celular/efectos de los fármacos , Cumarinas/química , Relación Dosis-Respuesta a Droga , Células HEK293 , Células Hep G2 , Humanos , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Imagen Óptica , Relación Estructura-Actividad
10.
Biosci Rep ; 39(1)2019 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-30602451

RESUMEN

This work tests bioenergetic and cell-biological implications of the synthetic fatty acid Minerval (2-hydroxyoleic acid), previously demonstrated to act by activation of sphingomyelin synthase in the plasma membrane (PM) and lowering of phosphatidylethanolamine (PE) and phosphatidylcholine (PC) and their carcinogenic signaling. We show here that Minerval also acts, selectively in cancer cell lines, as an ATP depleting uncoupler of mitochondrial oxidative phosphorylation (OxPhos). As a function of its exposure time, Minerval compromised the capacity of glioblastoma U87-MG cells to compensate for aberrant respiration by up-modulation of glycolysis. This effect was not exposure time-dependent in the lung carcinoma A549 cell line, which was more sensitive to Minerval. Compared with OxPhos inhibitors FCCP (uncoupler), rotenone (electron transfer inhibitor), and oligomycin (F1F0-ATPase inhibitor), Minerval action was similar only to that of FCCP. This similarity was manifested by mitochondrial membrane potential (MMP) depolarization, facilitation of oxygen consumption rate (OCR), restriction of mitochondrial and cellular reactive oxygen species (ROS) generation and mitochondrial fragmentation. Additionally, compared with other OxPhos inhibitors, Minerval uniquely induced ER stress in cancer cell lines. These new modes of action for Minerval, capitalizing on the high fatty acid requirements of cancer cells, can potentially enhance its cancer-selective toxicity and improve its therapeutic capacity.


Asunto(s)
Metabolismo Energético/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Ácidos Oléicos/farmacología , Células A549 , Adenosina Trifosfato/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Respiración de la Célula/efectos de los fármacos , Transporte de Electrón/efectos de los fármacos , Glucólisis/efectos de los fármacos , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Fosforilación Oxidativa/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
11.
Nat Commun ; 10(1): 62, 2019 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-30622276

RESUMEN

The extension of the amyloid hypothesis to include non-protein metabolite assemblies invokes a paradigm for the pathology of inborn error of metabolism disorders. However, a direct demonstration of the assembly of metabolite amyloid-like structures has so far been provided only in vitro. Here, we established an in vivo model of adenine self-assembly in yeast, in which toxicity is associated with intracellular accumulation of the metabolite. Using a strain blocked in the enzymatic pathway downstream to adenine, we observed a non-linear dose-dependent growth inhibition. Both the staining with an indicative amyloid dye and anti-adenine assemblies antibodies demonstrated the accumulation of adenine amyloid-like structures, which were eliminated by lowering the supplied adenine levels. Treatment with a polyphenol inhibitor reduced the occurrence of amyloid-like structures while not affecting the dramatic increase in intracellular adenine concentration, resulting in inhibition of cytotoxicity, further supporting the notion that toxicity is triggered by adenine assemblies.


Asunto(s)
Adenina/metabolismo , Amiloide/metabolismo , Errores Innatos del Metabolismo/etiología , Saccharomyces cerevisiae/metabolismo , Adenina/toxicidad , Amiloide/toxicidad , Errores Innatos del Metabolismo/metabolismo
12.
Exp Hematol ; 70: 97-108, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30593830

RESUMEN

Cell surface molecules aberrantly expressed or overexpressed by myeloid leukemic cells represent potential disease-specific therapeutic targets for antibodies. MUC1 is a polymorphic glycoprotein, the cleavage of which yields two unequal chains: a large extracellular α subunit containing a tandem repeat array bound in a strong noncovalent interaction to a smaller ß subunit containing the transmembrane and cytoplasmic domains. Because the α-chain can be released from the cell-bound domains of MUC1, agents directed against the α-chain will not effectively target MUC1+ cells. The MUC1 SEA (a highly conserved protein module so called from its initial identification in a sea urchin sperm protein, in enterokinase, and in agrin) domain formed by the binding of the α and ß chains  represents a stable structure fixed to the cell surface at all times. DMB-5F3, a partially humanized murine anti-MUC1 SEA domain monoclonal antibody, was used to examine MUC1 expression in acute myeloid leukemia (AML) and was found to bind acute myelomonocytic and monocytic leukemia (AML-M4 and AML-M5) cell lines. We also examined monocytic neoplasms freshly obtained from patients including chronic myelomonocytic leukemia and juvenile myelomonocytic leukemia, which were found to uniformly express MUC1. CD34+/lin-/CD38- or CD38+ presumed leukemic stem cell populations from CD34+ AML and CD34-CD38- or CD38+ populations from CD34- AML were also found to express MUC1, although at low percentages. Based on these studies, we generated an anti-MUC1 immunotoxin to directly gauge the cytotoxic efficacy of targeting AML-bound MUC1. Using single-chain DMB-5F3 fused to recombinant gelonin toxin, the degree of AML cytotoxicity was found to correlate with MUC1 expression. Our data support the use of an anti-MUC1 SEA module-drug conjugates to selectively target and inhibit MUC1-expressing myelomonocytic leukemic cells.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Sistemas de Liberación de Medicamentos , Inmunotoxinas/farmacología , Leucemia Mielomonocítica Crónica , Leucemia Mielomonocítica Juvenil , Mucina-1/inmunología , Proteínas de Neoplasias/inmunología , Proteínas Inactivadoras de Ribosomas Tipo 1/farmacología , Anticuerpos de Cadena Única/farmacología , Animales , Femenino , Humanos , Células K562 , Leucemia Mielomonocítica Crónica/tratamiento farmacológico , Leucemia Mielomonocítica Crónica/genética , Leucemia Mielomonocítica Crónica/inmunología , Leucemia Mielomonocítica Crónica/patología , Leucemia Mielomonocítica Juvenil/tratamiento farmacológico , Leucemia Mielomonocítica Juvenil/genética , Leucemia Mielomonocítica Juvenil/inmunología , Leucemia Mielomonocítica Juvenil/patología , Masculino , Ratones , Mucina-1/genética , Proteínas de Neoplasias/genética , Células Madre Neoplásicas
13.
Biochim Biophys Acta Gen Subj ; 1862(7): 1565-1575, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29634991

RESUMEN

BACKGROUND: Alzheimer's disease (AD) is the most common neurodegenerative disorder which is characterized by the deposits of intra-cellular tau protein and extra-cellular amyloid-ß (Aß) peptides in the human brain. Understanding the mechanism of protein aggregation and finding compounds that are capable of inhibiting its aggregation is considered to be highly important for disease therapy. METHODS: We used an in vitro High-Throughput Screening for the identification of potent inhibitors of tau aggregation using a proxy model; a highly aggregation-prone hexapeptide fragment 306VQIVYK311 derived from tau. Using ThS fluorescence assay we screened a library of 2401 FDA approved, bio-active and natural compounds in attempt to find molecules which can efficiently modulate tau aggregation. RESULTS: Among the screened compounds, palmatine chloride (PC) alkaloid was able to dramatically reduce the aggregation propensity of PHF6 at sub-molar concentrations. PC was also able to disassemble preformed aggregates of PHF6 and reduce the amyloid content in a dose-dependent manner. Insights obtained from MD simulation showed that PC interacted with the key residues of PHF6 responsible for ß-sheet formation, which could likely be the mechanism of inhibition and disassembly. Furthermore, PC could effectively inhibit the aggregation of full-length tau and disassemble preformed aggregates. CONCLUSIONS: We found that PC possesses "dual functionality" towards PHF6 and full-length tau, i.e. inhibit their aggregation and disassemble pre-formed fibrils. GENERAL SIGNIFICANCE: The "dual functionality" of PC is valuable as a disease modifying strategy for AD, and other tauopathies, by inhibiting their progress and reducing the effect of fibrils already present in the brain.


Asunto(s)
Alcaloides de Berberina/farmacología , Fragmentos de Péptidos/efectos de los fármacos , Proteínas tau/efectos de los fármacos , Neoplasias de las Glándulas Suprarrenales/patología , Amiloide/efectos de los fármacos , Amiloide/ultraestructura , Dicroismo Circular , Simulación por Computador , Ensayos Analíticos de Alto Rendimiento , Humanos , Técnicas In Vitro , Simulación del Acoplamiento Molecular , Fragmentos de Péptidos/química , Feocromocitoma/patología , Agregación Patológica de Proteínas , Células Tumorales Cultivadas , Proteínas tau/química
14.
Dis Model Mech ; 10(5): 645-654, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28213588

RESUMEN

Amyotrophic lateral sclerosis (ALS) is an incurable motor neurodegenerative disease caused by a diversity of genetic and environmental factors that leads to neuromuscular degeneration and has pathophysiological implications in non-neural systems. Our previous work showed abnormal levels of mRNA expression for biomarker genes in non-neuronal cell samples from ALS patients. The same genes proved to be differentially expressed in the brain, spinal cord and muscle of the SOD1G93A ALS mouse model. These observations support the idea that there is a pathophysiological relevance for the ALS biomarkers discovered in human mesenchymal stem cells (hMSCs) isolated from bone marrow samples of ALS patients (ALS-hMSCs). Here, we demonstrate that ALS-hMSCs are also a useful patient-based model to study intrinsic cell molecular mechanisms of the disease. We investigated the ALS-hMSC response to oxidative DNA damage exerted by neocarzinostatin (NCS)-induced DNA double-strand breaks (DSBs). We found that the ALS-hMSCs responded to this stress differently from cells taken from healthy controls (HC-hMSCs). Interestingly, we found that ALS-hMSC death in response to induction of DSBs was dependent on autophagy, which was initialized by an increase of phosphorylated (p)AMPK, and blocked by the class III phosphoinositide 3-kinase (PI3K) and autophagy inhibitor 3-methyladenine (3MeA). ALS-hMSC death in response to DSBs was not apoptotic as it was caspase independent. This unique ALS-hMSC-specific response to DNA damage emphasizes the possibility that an intrinsic abnormal regulatory mechanism controlling autophagy initiation exists in ALS-patient-derived hMSCs. This mechanism may also be relevant to the most-affected tissues in ALS. Hence, our approach might open avenues for new personalized therapies for ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Autofagia , Células de la Médula Ósea/metabolismo , Roturas del ADN de Doble Cadena , Células Madre Mesenquimatosas/metabolismo , Esclerosis Amiotrófica Lateral/genética , Humanos
15.
PLoS One ; 11(10): e0165031, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27768738

RESUMEN

Translation of mRNA in alternate reading frames (ARF) is a naturally occurring process heretofore underappreciated as a generator of protein diversity. The MUC1 gene encodes MUC1-TM, a signal-transducing trans-membrane protein highly expressed in human malignancies. Here we show that an AUG codon downstream to the MUC1-TM initiation codon initiates an alternate reading frame thereby generating a novel protein, MUC1-ARF. MUC1-ARF, like its MUC1-TM 'parent' protein, contains a tandem repeat (VNTR) domain. However, the amino acid sequence of the MUC1-ARF tandem repeat as well as N- and C- sequences flanking it differ entirely from those of MUC1-TM. In vitro protein synthesis assays and extensive immunohistochemical as well as western blot analyses with MUC1-ARF specific monoclonal antibodies confirmed MUC1-ARF expression. Rather than being expressed at the cell membrane like MUC1-TM, immunostaining showed that MUC1-ARF protein localizes mainly in the nucleus: Immunohistochemical analyses of MUC1-expressing tissues demonstrated MUC1-ARF expression in the nuclei of secretory luminal epithelial cells. MUC1-ARF expression varies in different malignancies. While the malignant epithelial cells of pancreatic cancer show limited expression, in breast cancer tissue MUC1-ARF demonstrates strong nuclear expression. Proinflammatory cytokines upregulate expression of MUC1-ARF protein and co-immunoprecipitation analyses demonstrate association of MUC1-ARF with SH3 domain-containing proteins. Mass spectrometry performed on proteins coprecipitating with MUC1-ARF demonstrated Glucose-6-phosphate 1-dehydrogenase (G6PD) and Dynamin 2 (DNM2). These studies not only reveal that the MUC1 gene generates a previously unidentified MUC1-ARF protein, they also show that just like its 'parent' MUC1-TM protein, MUC1-ARF is apparently linked to signaling and malignancy, yet a definitive link to these processes and the roles it plays awaits a precise identification of its molecular functions. Comprising at least 524 amino acids, MUC1-ARF is, furthermore, the longest ARF protein heretofore described.


Asunto(s)
Núcleo Celular/metabolismo , Mucina-1/genética , Biosíntesis de Proteínas , ARN Mensajero/genética , Animales , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Codón , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Ratones , Mucina-1/metabolismo , Neoplasias Pancreáticas/metabolismo
16.
Cancer Res ; 72(13): 3324-36, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22507854

RESUMEN

The cell-surface glycoprotein MUC1 is a particularly appealing target for antibody targeting, being selectively overexpressed in many types of cancers and a high proportion of cancer stem-like cells. However the occurrence of MUC1 cleavage, which leads to the release of the extracellular α subunit into the circulation where it can sequester many anti-MUC1 antibodies, renders the target problematic to some degree. To address this issue, we generated a set of unique MUC1 monoclonal antibodies that target a region termed the SEA domain that remains tethered to the cell surface after MUC1 cleavage. In breast cancer cell populations, these antibodies bound the cancer cells with high picomolar affinity. Starting with a partially humanized antibody, DMB5F3, we created a recombinant chimeric antibody that bound a panel of MUC1+ cancer cells with higher affinities relative to cetuximab (anti-EGFR1) or tratuzumab (anti-erbB2) control antibodies. DMB5F3 internalization from the cell surface occurred in an efficient temperature-dependent manner. Linkage to toxin rendered these DMB5F3 antibodies to be cytotoxic against MUC1+ cancer cells at low picomolar concentrations. Our findings show that high-affinity antibodies to cell-bound MUC1 SEA domain exert specific cytotoxicity against cancer cells, and they point to the SEA domain as a potential immunogen to generate MUC1 vaccines.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Mucina-1/inmunología , Western Blotting , Línea Celular Tumoral , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos
17.
Biochem Biophys Res Commun ; 414(4): 706-11, 2011 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-22001924

RESUMEN

We previously suggested a mechanism whereby the RNA induced silencing complex (RISC) brings about a specific cleavage at the sarcin-ricin loop (SRL) of 28S ribosomal RNA thereby eliciting translational suppression. Here we experimentally show that endogenous cleavages take place at the SRL site, in both mammalian cells and in Caenorhabditis elegans. Furthermore we demonstrate that bulged and looped-out residues present in the imperfect miRNA-[mRNA target site] duplexes, are complementary to the SRL site. These results support, and are compatible with, our described mechanism whereby microRNAs mediate cleavage of the highly conserved 28S rRNA sarcin/ricin loop leading to translational suppression.


Asunto(s)
MicroARNs/metabolismo , Biosíntesis de Proteínas , División del ARN , Ribosomas/metabolismo , Animales , Caenorhabditis elegans/enzimología , Caenorhabditis elegans/genética , Línea Celular Tumoral , Endorribonucleasas/química , Endorribonucleasas/metabolismo , Proteínas Fúngicas/química , Proteínas Fúngicas/metabolismo , Humanos , Ratones , MicroARNs/química , Conformación de Ácido Nucleico , ARN Ribosómico 28S/genética , ARN Ribosómico 28S/metabolismo
18.
Protein Sci ; 19(6): 1272-8, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20512980

RESUMEN

We report structural, functional, and biochemical similarities between Argonautes, the effector proteins of RNA-induced silencing complexes (RISCs), and alpha-sarcin-like ribotoxins. At the structural level, regions of similarity in the amino acid sequence are located in protein loops both in the ribotoxins and in the Argonautes. In ribotoxins, these protein loops confer specificity for a highly conserved segment of ribosomal RNA, the Sarcin-Ricin-Loop (SRL) that undergoes cleavage by the ribotoxin ribonuclease. This leads to suppression of translation. In addition to the structural similarity with ribotoxins, the Argonaute proteins (Ago) show both functional and biochemical parallels. Like the ribotoxins, the Agos exhibit ribonuclease activity and like the ribotoxins, translational suppression mediated by miRISC-resident Ago is accompanied by intact polysomes. Furthermore, in both translationally suppressed systems, the puromycin reaction, reflecting correct translocation and peptidyl-transferase activities, is unharmed. These findings support a mechanism for Ago-miRISCs whereby regulated cleavage of ribosomal RNA leads to translational suppression.


Asunto(s)
Endorribonucleasas/química , Factores Eucarióticos de Iniciación/química , Proteínas Fúngicas/química , Complejo Silenciador Inducido por ARN/química , Homología de Secuencia de Aminoácido , Secuencia de Aminoácidos , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Bovinos , Secuencia Conservada , Endorribonucleasas/metabolismo , Factores Eucarióticos de Iniciación/metabolismo , Proteínas Fúngicas/metabolismo , Humanos , Ratones , MicroARNs/química , MicroARNs/metabolismo , Modelos Genéticos , Modelos Moleculares , Datos de Secuencia Molecular , Biosíntesis de Proteínas , Estructura Terciaria de Proteína , Puromicina , Procesamiento Postranscripcional del ARN , ARN Ribosómico/metabolismo , Complejo Silenciador Inducido por ARN/metabolismo , Alineación de Secuencia
19.
Int J Cancer ; 124(1): 46-54, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-18821582

RESUMEN

MUC1, a heavily glycosylated mucin, has generated considerable interest as a target for tumor killing because of its overexpression in malignancies. Full-length MUC1 (MUC1/TM) is proteolytically cleaved after synthesis generating alpha and beta subunits, which specifically bind in a noncovalent interaction. Although the beta chain remains on the cell surface, the alpha chain binds in an on-and-off interaction. Most anti-MUC1 antibodies (Abs) described to date recognize epitopes within the highly immunogenic alpha-chain tandem repeat. Because the alpha-chain is shed, such Abs are sequestered and fail to reach MUC1-expressing cells. Immunizing with cDNA encoding MUC1/TM and the spliced MUC1/X isoform from which the tandem repeat has been deleted yielded antibodies to the MUC1 alpha/beta junction. Pseudomonas toxin PE38 linked to polyclonal anti-MUC1 alpha/beta junction Abs both bound and killed MUC1-positive malignant cells. Monoclonal DMC209 binds the MUC1 alpha/beta junction in both MUC1/X and MUC1/TM. When injected into SCID mice xenotransplanted with human breast cancer MDA-MB-231, monoclonal DMC209 showed significant in vivo tumor-suppressive activity. The MUC1/X alpha/beta junction presents a biologically-significant target in MUC1-expressing malignancies because (i) antibodies directed against cell-bound alpha/beta junction epitopes reach the intended cellular target, (ii) antibodies to junction epitope are internalized into cells, (iii) anti alpha/beta junction antibodies can effectively kill high MUC1-expressing cancer cells as antibody-toxin conjugates and (iv) antibodies targeting the MUC1 cell-bound alpha/beta junction results in tumor suppression in vivo. Our results indicate that cell-bound MUC1 alpha/beta junction, unlike shed alpha chain, represents a highly effective moiety for targeting and killing MUC1-expressing malignancies.


Asunto(s)
Inmunoterapia/métodos , Inmunotoxinas/química , Mucina-1/fisiología , Animales , Anticuerpos Monoclonales/química , Epítopos/química , Femenino , Humanos , Hibridomas/metabolismo , Ratones , Ratones SCID , Mucina-1/metabolismo , Trasplante de Neoplasias , Conformación Proteica , Isoformas de Proteínas , Estructura Terciaria de Proteína
20.
J Biol Chem ; 283(24): 16928-39, 2008 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-18387948

RESUMEN

We report here syntenic loci in humans and mice incorporating gene clusters coding for secreted proteins each comprising 10 cysteine residues. These conform to three-fingered protein/Ly-6/urokinase-type plasminogen activator receptor (uPAR) domains that shape three-fingered proteins (TFPs). The founding gene is PATE, expressed primarily in prostate and less in testis. We have identified additional human PATE-like genes (PATE-M, PATE-DJ, and PATE-B) that co-localize with the PATE locus, code for novel secreted PATE-like proteins, and show selective expression in prostate and/or testis. Anti-PATE-B-specific antibodies demonstrated the presence of PATE-B in the region of the sperm acrosome and at high levels on malignant prostatic epithelial cells. The syntenic mouse Pate-like locus encompasses 14 active genes coding for secreted proteins, which are all, except for Pate-P and Pate-Q, expressed primarily in prostate and/or testis. Pate-P and Pate-Q are expressed solely in placental tissue. Castration up-regulates prostate expression of mouse Pate-B and Pate-E, whereas testosterone ablates this induced expression. The sequence similarity between TFP/Ly-6/uPAR proteins that modulate activity of nicotinic acetylcholine receptors and the PATE (Pate)-like proteins stimulated us to see whether these proteins possess analogous activity. Pharmacological studies showed significant modulation of the nicotinic acetylcholines by the PATE-B, Pate-C, and Pate-P proteins. In concert with these findings, certain PATE (Pate)-like genes were extensively expressed in neuron-rich tissues. Taken together, our findings indicate that in addition to participation of the PATE (Pate)-like genes in functions related to fertility and reproduction, some of them likely act as important modulators of neural transmission.


Asunto(s)
Antígenos Ly/genética , Proteínas de la Membrana/genética , Familia de Multigenes , Neuronas/metabolismo , Receptores de Superficie Celular/genética , Sistema Urogenital/metabolismo , Secuencia de Aminoácidos , Animales , Femenino , Humanos , Masculino , Ratones , Datos de Secuencia Molecular , Oocitos/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Homología de Secuencia de Aminoácido , Distribución Tisular , Xenopus laevis/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...