Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 154
Filtrar
1.
Mar Drugs ; 22(4)2024 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-38667791

RESUMEN

Cholestane-3ß,5α,6ß-triol (CT) and its analogues are abundant in natural sources and are reported to demonstrate cytotoxicity toward different kinds of tumor cells without a deep probe into their mechanism of action. CT is also one of the major metabolic oxysterols of cholesterol in mammals and is found to accumulate in various diseases. An extensive exploration of the biological roles of CT over the past few decades has established its identity as an apoptosis inducer. In this study, the effects of CT on A549 cell death were investigated through cell viability assays. RNA-sequencing analysis and western blot of CT-treated A549 cells revealed the role of CT in inducing endoplasmic reticulum (ER) stress response and enhancing autophagy flux, suggesting a putative mechanism of CT-induced cell-death activation involving reactive oxygen species (ROS)-mediated ER stress and autophagy. It is reported for the first time that the upregulation of autophagy induced by CT can serve as a cellular cytotoxicity response in accelerating CT-induced cell death in A549 cells. This research provides evidence for the effect of CT as an oxysterol in cell response to oxidative damage and allows for a deep understanding of cholesterol in its response in an oxidative stress environment that commonly occurs in the progression of various diseases.


Asunto(s)
Autofagia , Supervivencia Celular , Colestanoles , Estrés del Retículo Endoplásmico , Especies Reactivas de Oxígeno , Humanos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Autofagia/efectos de los fármacos , Células A549 , Especies Reactivas de Oxígeno/metabolismo , Supervivencia Celular/efectos de los fármacos , Apoptosis/efectos de los fármacos , Colesterol/metabolismo , Colestanos/farmacología , Muerte Celular/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos
2.
FASEB J ; 36(12): e22655, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36421008

RESUMEN

Trodusquemine is an aminosterol with a variety of biological and pharmacological functions, such as acting as an antimicrobial, stimulating body weight loss and interfering with the toxicity of proteins involved in the development of Alzheimer's and Parkinson's diseases. The mechanisms of interaction of aminosterols with cells are, however, still largely uncharacterized. Here, by using fluorescently labeled trodusquemine (TRO-A594 and TRO-ATTO565), we show that trodusquemine binds initially to the plasma membrane of living cells, that the binding affinity is dependent on cholesterol, and that trodusquemine is then internalized and mainly targeted to lysosomes after internalization. We also found that TRO-A594 is able to strongly and selectively bind to myelinated fibers in fixed mouse brain slices, and that it is a marker compatible with tissue clearing and light-sheet fluorescence microscopy or expansion microscopy. In conclusion, this work contributes to further characterize the biology of aminosterols and provides a new tool for nerve labeling suitable for the most advanced microscopy techniques.


Asunto(s)
Colestanos , Animales , Ratones , Colestanos/farmacología , Espermina/farmacología , Microscopía Fluorescente/métodos , Colesterol
3.
Fitoterapia ; 158: 105174, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35296434

RESUMEN

Five new cholestane glycosides, named parisfargosides A-E (1-5), were isolated from the rhizomes of Paris fargesii. Their structures were elucidated on the basis of UV, HR-ESI-MS, 1D and 2D NMR data as well as chemical methods. The structures of all compounds contained α, ß-unsaturated ketone unit. Compounds 3-5 possessed a 16,23-cyclocholest skeleton with 6/6/6/5/5 condensed ring, and the absolute configurations of C-16 and C-23 were confirmed according to ROESY spectra with pyridine­d5 and DMSO­d6 as solvents. In addition, the platelet aggregation activity and cytotoxic activity against five human cancer cell lines (HL-60, A549, SMMC-7721, MDA-MB-231, and SW480) of compounds 1-5 were evaluated.


Asunto(s)
Colestanos , Liliaceae , Colestanos/farmacología , Glicósidos/química , Humanos , Estructura Molecular , Rizoma/química
4.
Am J Physiol Cell Physiol ; 321(3): C569-C584, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34288720

RESUMEN

Rheumatoid arthritis (RA) is a debilitating autoimmune disease of unknown cause, characterized by infiltration and accumulation of activated immune cells in the synovial joints where cartilage and bone destructions occur. Myeloid-derived suppressor cells (MDSCs) are of myeloid origin and are able to suppress T cell responses. Src homology 2 domain-containing inositol polyphosphate 5-phosphatase 1 (SHIP1) was shown to be involved in the regulation of MDSC differentiation. The purpose of the present study was to investigate the effect of inhibition of SHIP1 on the expansion of MDSCs in RA using a collagen-induced inflammatory arthritis (CIA) mouse model. In DBA/1 mice, treatment with a small molecule-specific SHIP1 inhibitor 3α-aminocholestane (3AC) induced a marked expansion of MDSCs in vivo. Both pretreatment with 3AC of DBA/1 mice prior to CIA induction and intervention with 3AC during CIA progression significantly reduced disease incidence and severity. Adoptive transfer of MDSCs isolated from 3AC-treated mice, but not naïve MDSCs from normal mice, into CIA mice significantly reduced disease incidence and severity, indicating that the 3AC-induced MDSCs were the cellular mediators of the observed amelioration of the disease. In conclusion, inhibition of SHIP1 expands MDSCs in vivo and attenuates development of CIA in mice. Small molecule-specific inhibition of SHIP1 may therefore offer therapeutic benefit to patients with RA and other autoimmune diseases.


Asunto(s)
Antiinflamatorios/farmacología , Artritis Experimental/tratamiento farmacológico , Colestanos/farmacología , Células Supresoras de Origen Mieloide/inmunología , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/genética , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Artritis Experimental/genética , Artritis Experimental/inmunología , Artritis Experimental/patología , Artritis Reumatoide/genética , Artritis Reumatoide/inmunología , Artritis Reumatoide/patología , Comunicación Celular , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Expresión Génica , Humanos , Cápsula Articular/efectos de los fármacos , Cápsula Articular/inmunología , Cápsula Articular/patología , Ratones , Ratones Endogámicos DBA , Ratones Noqueados , Células Supresoras de Origen Mieloide/citología , Células Supresoras de Origen Mieloide/trasplante , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/antagonistas & inhibidores , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas/inmunología , Índice de Severidad de la Enfermedad , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/patología
5.
J Biol Chem ; 297(1): 100818, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34029592

RESUMEN

The cleavage of the insulin receptor by ß-secretase 1 (BACE1) in the liver increases during diabetes, which contributes to reduce insulin receptor levels and impair insulin signaling. However, the precise signaling events that lead to this increased cleavage are unclear. We showed that BACE1 cleaves the insulin receptor in the early secretory pathway. Indeed, coimmunoprecipitation experiments reveal the interaction of the proforms of the two proteins. Moreover, fragments of insulin receptor are detected in the early secretory pathway and a mutated form of BACE1 that retains its prodomain cleaves an early secretory pathway-resident form of the insulin receptor. We showed that BACE1 proform levels are regulated by proteasome and/or lysosome-dependent degradation systems whose efficiencies are dependent on the O-GlcNacylation process. Our results showed that enhanced O-GlcNacylation reduces the efficiency of intracellular protein degradation systems, leading to the accumulation of the proform of BACE1 in the early secretory pathway where it cleaves the precursor of the insulin receptor. All these dysregulations are found in the livers of diabetic mice. In addition, we performed a screen of molecules according to their ability to increase levels of the insulin receptor at the surface of BACE1-overexpressing cells. This approach identified the aminosterol Claramine, which accelerated intracellular trafficking of the proform of BACE1 and increased autophagy. Both of these effects likely contribute to the reduced amount of the proform of BACE1 in the early secretory pathway, thereby reducing insulin receptor cleavage. These newly described properties of Claramine are consistent with its insulin sensitizing effect.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Colestanos/farmacología , Receptor de Insulina/metabolismo , Espermina/análogos & derivados , Animales , Diabetes Mellitus Experimental/patología , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico/efectos de los fármacos , Glicosilación/efectos de los fármacos , Células HEK293 , Células Hep G2 , Humanos , Hígado/patología , Modelos Biológicos , Unión Proteica/efectos de los fármacos , Proteolisis/efectos de los fármacos , Proteostasis/efectos de los fármacos , Vías Secretoras/efectos de los fármacos , Espermina/farmacología , Ubiquitina/metabolismo , Ubiquitinación/efectos de los fármacos
6.
Neurobiol Dis ; 156: 105402, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34044147

RESUMEN

Mutations in the beta-amyloid protein (APP) cause familial Alzheimer's disease. In hAPP-J20 mice expressing mutant APP, pharmacological inhibition or genetic ablation of the tyrosine phosphatase PTP1B prevents CA3 hippocampus neuron loss and cognitive decline. However, how targeting PTP1B affects the cellular mechanisms underlying these cognitive deficits remains unknown. Changes in synaptic strength at the hippocampus can affect information processing for learning and memory. While prior studies have focused on post-synaptic mechanisms to account for synaptic deficits in Alzheimer's disease models, presynaptic mechanisms may also be affected. Here, using whole cell patch-clamp recording, coefficient of variation (CV) analysis suggested a profound presynaptic deficit in long-term potentiation (LTP) of CA3:CA1 synapses in hAPP-J20 mice. While the membrane-impermeable ionotropic NMDA receptor (NMDAR) blocker norketamine in the post-synaptic recording electrode had no effect on LTP, additional bath application of the ionotropic NMDAR blockers MK801 could replicate the deficit in LTP in wild type mice. In contrast to LTP, the paired-pulse ratio and short-term facilitation (STF) were aberrantly increased in hAPP-J20 mice. These synaptic deficits in hAPP-J20 mice were associated with reduced phosphorylation of NMDAR GluN2B and the synaptic vesicle recycling protein NSF (N-ethylmaleimide sensitive factor). Phosphorylation of both proteins, together with synaptic plasticity and cognitive function, were restored by PTP1B ablation or inhibition by the PTP1B-selective inhibitor Trodusquemine. Taken together, our results indicate that PTP1B impairs presynaptic NMDAR-mediated synaptic plasticity required for spatial learning in a mouse model of Alzheimer's disease. Since Trodusquemine has undergone phase 1/2 clinical trials to treat obesity, it could be repurposed to treat Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Plasticidad Neuronal/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores Presinapticos/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Animales , Colestanos/farmacología , Colestanos/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Plasticidad Neuronal/efectos de los fármacos , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Receptores de N-Metil-D-Aspartato/genética , Receptores Presinapticos/genética , Espermina/análogos & derivados , Espermina/farmacología , Espermina/uso terapéutico
7.
Neurobiol Dis ; 155: 105397, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34015491

RESUMEN

Subanesthetic doses of ketamine induce schizophrenia-like behaviors in mice including hyperlocomotion and deficits in working memory and sensorimotor gating. Here, we examined the effect of in vivo ketamine administration on neuronal properties and endocannabinoid (eCB)-dependent modulation of synaptic transmission onto layer 2/3 pyramidal neurons in brain slices of the prefrontal cortex, a region tied to the schizophrenia-like behavioral phenotypes of ketamine. Since deficits in working memory and sensorimotor gating are tied to activation of the tyrosine phosphatase PTP1B in glutamatergic neurons, we asked whether PTP1B contributes to these effects of ketamine. Ketamine increased membrane resistance and excitability of pyramidal neurons. Systemic pharmacological inhibition of PTP1B by Trodusquemine restored these neuronal properties and prevented each of the three main ketamine-induced behavior deficits. Ketamine also reduced mobilization of eCB by pyramidal neurons, while unexpectedly reducing their inhibitory inputs, and these effects of ketamine were blocked or occluded by PTP1B ablation in glutamatergic neurons. While ablation of PTP1B in glutamatergic neurons prevented ketamine-induced deficits in memory and sensorimotor gating, it failed to prevent hyperlocomotion (a psychosis-like phenotype). Taken together, these results suggest that PTP1B in glutamatergic neurons mediates ketamine-induced deficits in eCB mobilization, memory and sensorimotor gating whereas PTP1B in other cell types contributes to hyperlocomotion. Our study suggests that the PTP1B inhibitor Trodusquemine may represent a new class of fast-acting antipsychotic drugs to treat schizophrenia-like symptoms.


Asunto(s)
Colestanos/farmacología , Ketamina/toxicidad , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Esquizofrenia/inducido químicamente , Esquizofrenia/prevención & control , Espermina/análogos & derivados , Proteínas Adaptadoras Transductoras de Señales/farmacología , Anestésicos Disociativos/toxicidad , Animales , Colestanos/uso terapéutico , Relación Dosis-Respuesta a Droga , Proteínas con Dominio LIM/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Esquizofrenia/metabolismo , Espermina/farmacología , Espermina/uso terapéutico
8.
Commun Biol ; 3(1): 435, 2020 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-32792544

RESUMEN

The onset and progression of numerous protein misfolding diseases are associated with the presence of oligomers formed during the aberrant aggregation of several different proteins, including amyloid-ß (Aß) in Alzheimer's disease and α-synuclein (αS) in Parkinson's disease. These small, soluble aggregates are currently major targets for drug discovery. In this study, we show that trodusquemine, a naturally-occurring aminosterol, markedly reduces the cytotoxicity of αS, Aß and HypF-N oligomers to human neuroblastoma cells by displacing the oligomers from cell membranes in the absence of any substantial morphological and structural changes to the oligomers. These results indicate that the reduced toxicity results from a mechanism that is common to oligomers from different proteins, shed light on the origin of the toxicity of the most deleterious species associated with protein aggregation and suggest that aminosterols have the therapeutically-relevant potential to protect cells from the oligomer-induced cytotoxicity associated with numerous protein misfolding diseases.


Asunto(s)
Membrana Celular/metabolismo , Colestanos/farmacología , Pliegue de Proteína , Multimerización de Proteína , Espermina/análogos & derivados , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/toxicidad , Fenómenos Biofísicos/efectos de los fármacos , Transferasas de Carboxilo y Carbamoilo/química , Transferasas de Carboxilo y Carbamoilo/toxicidad , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/toxicidad , Humanos , Pliegue de Proteína/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Espermina/farmacología , alfa-Sinucleína/química , alfa-Sinucleína/toxicidad
9.
J Nat Prod ; 83(4): 1043-1050, 2020 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-32227943

RESUMEN

An extract of Galtonia regalis from the Natural Products Discovery Institute showed moderate antiplasmodial activity, with an IC50 value less than 1.25 µg/mL. The two known cholestane glycosides 1 and 2 and the five new cholestane glycosides galtonosides A-E (3-7) were isolated after bioassay-directed fractionation. The structures of the new compounds were determined by interpretation of their NMR and mass spectra. Among these compounds, galtonoside B (4) displayed the most potent antiplasmodial activity, with an IC50 value of 0.214 µM against the drug-resistant Dd2 strain of Plasmodium falciparum.


Asunto(s)
Antimaláricos/química , Colestanos/farmacología , Glicósidos/farmacología , Asparagales/química , Colestanos/química , Colestanos/aislamiento & purificación , Glicósidos/química , Glicósidos/aislamiento & purificación , Humanos , Extractos Vegetales/química , Extractos Vegetales/aislamiento & purificación , Plasmodium falciparum/química
10.
J Neurosci ; 40(7): 1581-1593, 2020 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-31915254

RESUMEN

Alzheimer's disease (AD) is the most common neurodegenerative disorder, resulting in the progressive decline of cognitive function in patients. Familial forms of AD are tied to mutations in the amyloid precursor protein, but the cellular mechanisms that cause AD remain unclear. Inflammation and amyloidosis from amyloid ß (Aß) aggregates are implicated in neuron loss and cognitive decline. Inflammation activates the protein-tyrosine phosphatase 1B (PTP1B), and this could suppress many signaling pathways that activate glycogen synthase kinase 3ß (GSK3ß) implicated in neurodegeneration. However, the significance of PTP1B in AD pathology remains unclear. Here, we show that pharmacological inhibition of PTP1B with trodusquemine or selective ablation of PTP1B in neurons prevents hippocampal neuron loss and spatial memory deficits in a transgenic AD mouse model with Aß pathology (hAPP-J20 mice of both sexes). Intriguingly, while systemic inhibition of PTP1B reduced inflammation in the hippocampus, neuronal PTP1B ablation did not. These results dissociate inflammation from neuronal loss and cognitive decline and demonstrate that neuronal PTP1B hastens neurodegeneration and cognitive decline in this model of AD. The protective effect of PTP1B inhibition or ablation coincides with the restoration of GSK3ß inhibition. Neuronal ablation of PTP1B did not affect cerebral amyloid levels or plaque numbers, but reduced Aß plaque size in the hippocampus. In summary, our preclinical study suggests that targeting PTP1B may be a new strategy to intervene in the progression of AD.SIGNIFICANCE STATEMENT Familial forms of Alzheimer's disease (AD) are tied to mutations in the amyloid precursor protein, but the cellular mechanisms that cause AD remain unclear. Here, we used a mouse model expressing human amyloid precursor protein bearing two familial mutations and asked whether activation of a phosphatase PTP1B participates in the disease process. Systemic inhibition of this phosphatase using a selective inhibitor prevented cognitive decline, neuron loss in the hippocampus, and attenuated inflammation. Importantly, neuron-targeted ablation of PTP1B also prevented cognitive decline and neuron loss but did not reduce inflammation. Therefore, neuronal loss rather than inflammation was critical for AD progression in this mouse model, and that disease progression could be ameliorated by inhibition of PTP1B.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Proteínas del Tejido Nervioso/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/fisiología , Memoria Espacial/fisiología , Péptidos beta-Amiloides/análisis , Animales , Colestanos/farmacología , Modelos Animales de Enfermedad , Femenino , Glucógeno Sintasa Quinasa 3 beta/fisiología , Hipocampo/efectos de los fármacos , Hipocampo/patología , Humanos , Inflamación , Resistencia a la Insulina , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Fragmentos de Péptidos/análisis , Placa Amiloide/patología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Proteínas Recombinantes/metabolismo , Memoria Espacial/efectos de los fármacos , Espermina/análogos & derivados , Espermina/farmacología
11.
Sci Rep ; 9(1): 18919, 2019 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-31831796

RESUMEN

Potentiating neuritogenesis through pharmacological intervention might hold therapeutic promise in neurodegenerative disorders and acute brain injury. Here, we investigated the novel neuritogenic potentials of a steroidal chlorohydrin, 3ß, 6ß-dichloro-5-hydroxy-5α-cholestane (hereafter, SCH) and the change in cellular proteome to gain insight into the underlying mechanism of its neurotrophic activity in hippocampal neurons. Morphometric analysis showed that SCH promoted early neuronal differentiation, dendritic arborization and axonal maturation. Proteomic and bioinformatic analysis revealed that SCH induced upregulation of several proteins, including those associated with neuronal differentiation and development. Immunocytochemical data further indicates that SCH-treated neurons showed upregulation of Hnrnpa2b1 and Map1b, validating their proteomic profiles. In addition, a protein-protein interaction network analysis identified TrkA as a potential target connecting most of the upregulated proteins. The neurite outgrowth effect of SCH was suppressed by TrkA inhibitor, GW441756, verifying TrkA-dependent activity of SCH, which further supports the connection of TrkA with the upregulated proteins. Also, the computational analysis revealed that SCH interacts with the NGF-binding domain of TrkA through Phe327 and Asn355. Collectively, our findings provide evidence that SCH promotes neuronal development via upregulating TrkA-signaling proteins and suggest that SCH could be a promising therapeutic agent in the prevention and treatment of neurodegenerative disorders.


Asunto(s)
Axones/metabolismo , Diferenciación Celular/efectos de los fármacos , Colestanos/farmacología , Dendritas/metabolismo , Hipocampo/metabolismo , Receptor trkA/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Axones/patología , Colestanos/química , Dendritas/patología , Hipocampo/patología , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba/efectos de los fármacos
12.
Phytochemistry ; 164: 206-214, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31177053

RESUMEN

Eight undescribed cholestane glycosides named osaundersioside A-H, along with three previously known compounds named osaundersioside I-K were isolated from Ornithogalum saundersiae Baker bulbs (Asparagaceae). Their structures were elucidated by extensive spectroscopic analysis and chemical methods. All isolates were evaluated for their cytotoxic activity and inhibitory effects on lipopolysaccharide (LPS)-induced nitric oxide (NO) production. Osaundersioside C was thus determined to exhibit specific cytotoxicity towards MCF-7 cell line with an IC50 value of 0.20 µM, Osaundersioside H exhibited inhibitory effect on NO production in macrophages at the concentration of 10-5 M, with inhibition rate of 56.81%.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Antineoplásicos Fitogénicos/farmacología , Asparagaceae/química , Colestanos/farmacología , Glicósidos/farmacología , Animales , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/aislamiento & purificación , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/aislamiento & purificación , Proliferación Celular/efectos de los fármacos , Colestanos/química , Colestanos/aislamiento & purificación , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Glicósidos/química , Glicósidos/aislamiento & purificación , Humanos , Lipopolisacáridos/antagonistas & inhibidores , Lipopolisacáridos/farmacología , Células MCF-7 , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Estructura Molecular , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/biosíntesis , Corteza de la Planta/química , Raíces de Plantas/química , Relación Estructura-Actividad
13.
Eur J Med Chem ; 168: 78-86, 2019 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-30798054

RESUMEN

22-Oxocholestanes bearing the oxime functionality in the side chain have been synthesized from diosgenin and evaluated in vivo as anti-inflammatory agents in an acute inflammation mouse ear model, against the commercial glucocorticoid dexamethasone. The final compounds were all regioselectively obtained with an E configuration at the oxime double bond. The title compounds reduced ear-induced inflammation and edema. The most active oximes repressed the expression of proinflammatory genes TNF-α, COX-2, and IL-6; including macrophage migration inhibitory factor. Overall, our data suggest that 22-oxocholestane oximes exert a strong in vivo anti-inflammatory activity.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Colestanos/farmacología , Enfermedades del Oído/tratamiento farmacológico , Edema/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Oximas/farmacología , Animales , Antiinflamatorios no Esteroideos/síntesis química , Antiinflamatorios no Esteroideos/química , Colestanos/síntesis química , Colestanos/química , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Enfermedades del Oído/metabolismo , Edema/metabolismo , Inflamación/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Estructura Molecular , Oximas/síntesis química , Oximas/química , Relación Estructura-Actividad , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
14.
Nat Commun ; 10(1): 225, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30644384

RESUMEN

Transient oligomeric species formed during the aggregation process of the 42-residue form of the amyloid-ß peptide (Aß42) are key pathogenic agents in Alzheimer's disease (AD). To investigate the relationship between Aß42 aggregation and its cytotoxicity and the influence of a potential drug on both phenomena, we have studied the effects of trodusquemine. This aminosterol enhances the rate of aggregation by promoting monomer-dependent secondary nucleation, but significantly reduces the toxicity of the resulting oligomers to neuroblastoma cells by inhibiting their binding to the cellular membranes. When administered to a C. elegans model of AD, we again observe an increase in aggregate formation alongside the suppression of Aß42-induced toxicity. In addition to oligomer displacement, the reduced toxicity could also point towards an increased rate of conversion of oligomers to less toxic fibrils. The ability of a small molecule to reduce the toxicity of oligomeric species represents a potential therapeutic strategy against AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/metabolismo , Colestanos/uso terapéutico , Fragmentos de Péptidos/metabolismo , Espermina/análogos & derivados , Péptidos beta-Amiloides/efectos de los fármacos , Animales , Caenorhabditis elegans , Línea Celular Tumoral , Colestanos/farmacología , Evaluación Preclínica de Medicamentos , Fragmentos de Péptidos/efectos de los fármacos , Espermina/farmacología , Espermina/uso terapéutico
15.
Fitoterapia ; 130: 241-246, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30196076

RESUMEN

Three new cholestane-type sterols bearing an unusual ∆22-24-oxo side chain, namely, dictyoptesterols A-C (1-3), were isolated from the brown alga Dictyopteris undulata Holmes, together with five known strutural analogues (4-8). Their structures were elucidated on the basis of by extensive spectroscopic analysis. The absolute configurations of the steroidal nuclei of the new compounds were proposed by a comparison of NMR data with those of related known compounds as well as biogenetic considerations. All of the isolates were evaluated in vitro for their potential to inhibit protein tyrosine phosphatase-1B (PTP1B) activity. The results showed that compounds 1-5 exhibited different levels of PTP1B inhibitory activities with IC50 values ranging from 3.03 ±â€¯0.76 to 15.01 ±â€¯2.88 µM. In particular, compounds 3 and 4 showed promising inhibitory effects towards PTP1B with IC50 values of 3.03 ±â€¯0.76 and 3.72 ±â€¯0.40 µM, respectively, when compared to the positive control oleanolic acid (IC50, 2.83 ±â€¯0.39 µM). The chemotaxonomic significance of these isolated ∆22-24-oxo cholestanes has also been discussed.


Asunto(s)
Colestanos/aislamiento & purificación , Inhibidores Enzimáticos/aislamiento & purificación , Phaeophyceae/química , Fitosteroles/aislamiento & purificación , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , China , Colestanos/farmacología , Inhibidores Enzimáticos/farmacología , Estructura Molecular , Fitosteroles/farmacología
16.
Vascul Pharmacol ; 109: 36-44, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29894845

RESUMEN

Protein tyrosine phosphatase 1B (PTP1B) impairs nitric oxide (NO) production and induces endothelial dysfunction in various diseases, including diabetes, septic shock and heart failure. In non-cardiovascular tissues, PTP1B modulates endoplasmic reticulum stress (ERS) however this role has never been assessed in endothelial cells. We evaluated the link between PTP1B, ERS and endothelial dysfunction in mice. Induction of ERS (Tunicamycin) in vivo in mice or ex vivo in mouse arteries led to severe arterial endothelial dysfunction (i.e. reduced flow-dependent, NO mediated dilatation in isolated small mesenteric arteries), and this was prevented by the PTP1B inhibitor trodusquemine and absent in PTP1B-/- mice. Trodusquemine also prevented the Tunicamycin -induced increased arterial levels of the molecular ERS actors 78 kDa glucose-regulated protein (GRP78) and Activating Transcription Factor 6 (ATF6α). Tunicamycin strongly increased the interactions of PTP1B with GRP78 and the activated forms of protein kinase RNA-like endoplasmic reticulum kinase (PERK) and IRE1α (proximity Ligation Assay). Thus, PTP1B plays a central role in the regulation of ERS in the endothelium, and the endothelial protective effect of PTP1B inhibition appears likely due at least in part to reduction of endothelial ERS, notably by promoting PERK protective pathway. Modulation of ER stress via PTP1B inhibitors may be a promising approach to protect the endothelium in cardiovascular diseases.


Asunto(s)
Estrés del Retículo Endoplásmico/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Tunicamicina/farmacología , Vasodilatación/efectos de los fármacos , Factor de Transcripción Activador 6/metabolismo , Animales , Colestanos/farmacología , Chaperón BiP del Retículo Endoplásmico , Endorribonucleasas/metabolismo , Endotelio Vascular/enzimología , Endotelio Vascular/fisiopatología , Inhibidores Enzimáticos/farmacología , Proteínas de Choque Térmico/metabolismo , Masculino , Ratones Endogámicos BALB C , Ratones Noqueados , Óxido Nítrico/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 1/deficiencia , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Transducción de Señal/efectos de los fármacos , Espermina/análogos & derivados , Espermina/farmacología , eIF-2 Quinasa/metabolismo
17.
ACS Chem Biol ; 13(8): 2308-2319, 2018 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-29953201

RESUMEN

The aggregation of α-synuclein, an intrinsically disordered protein that is highly abundant in neurons, is closely associated with the onset and progression of Parkinson's disease. We have shown previously that the aminosterol squalamine can inhibit the lipid induced initiation process in the aggregation of α-synuclein, and we report here that the related compound trodusquemine is capable of inhibiting not only this process but also the fibril-dependent secondary pathways in the aggregation reaction. We further demonstrate that trodusquemine can effectively suppress the toxicity of α-synuclein oligomers in neuronal cells, and that its administration, even after the initial growth phase, leads to a dramatic reduction in the number of α-synuclein inclusions in a Caenorhabditis elegans model of Parkinson's disease, eliminates the related muscle paralysis, and increases lifespan. On the basis of these findings, we show that trodusquemine is able to inhibit multiple events in the aggregation process of α-synuclein and hence to provide important information about the link between such events and neurodegeneration, as it is initiated and progresses. Particularly in the light of the previously reported ability of trodusquemine to cross the blood-brain barrier and to promote tissue regeneration, the present results suggest that this compound has the potential to be an important therapeutic candidate for Parkinson's disease and related disorders.


Asunto(s)
Colestanos/farmacología , Enfermedad de Parkinson/tratamiento farmacológico , Agregado de Proteínas/efectos de los fármacos , Agregación Patológica de Proteínas/prevención & control , Espermina/análogos & derivados , alfa-Sinucleína/metabolismo , Animales , Caenorhabditis elegans/fisiología , Línea Celular , Colestanos/uso terapéutico , Modelos Animales de Enfermedad , Humanos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Enfermedad de Parkinson/metabolismo , Agregación Patológica de Proteínas/metabolismo , Espermina/farmacología , Espermina/uso terapéutico
18.
ChemMedChem ; 13(10): 1018-1027, 2018 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-29465814

RESUMEN

The emergence of multidrug-resistant bacteria and pathogens has created an urgent need for the development of new antibiotics. Herein we report our investigations into the broad-spectrum activity of an easily prepared water-soluble polyaminosterol compound, namely claramine A1, against both drug-sensitive and drug-resistant Gram-negative and Gram-positive bacterial strains. We also report its peculiar mechanism of action, which differs from that of all the other well-known classes of antibiotics, toward Gram-negative and Gram-positive bacteria. Given their low cytotoxicity, this class of compounds based on claramine A1 could constitute an effective response to combat the emergence of multidrug-resistant bacteria and nosocomial diseases.


Asunto(s)
Antiinfecciosos/química , Antiinfecciosos/farmacología , Colestanos/química , Colestanos/farmacología , Espermina/análogos & derivados , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Humanos , Larva/efectos de los fármacos , Pruebas de Micronúcleos , Estructura Molecular , Mariposas Nocturnas/efectos de los fármacos , Espermina/química , Espermina/farmacología , Relación Estructura-Actividad
19.
Molecules ; 22(8)2017 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-28757596

RESUMEN

Previous phytochemical studies of the bulbs of Ornithogalum saundersiae, an ornamental perennial plant native to South Africa, resulted in the isolation of 29 new cholestane glycosides, some of which were structurally unique and showed potent cytotoxic activity against cultured tumor cell lines. Therefore, we aimed to perform further phytochemical examinations of methanolic extracts obtained from Ornithogalum saundersiae bulbs, isolating 12 new cholestane rhamnosides (1-12) and seven known compounds (13-19). The structures of the new compounds (1-12) were identified via NMR-based structural characterization methods, and through a sequence of chemical transformations followed by spectroscopic and chromatographic analysis. The cytotoxic activity of the isolated compounds (1-19) and the derivatives (1a and 6a) against HL-60 human promyelocytic leukemia cells and A549 human lung adenocarcinoma cells was evaluated. Compounds 10-12, 16, and 17 showed cytotoxicity against both HL-60 and A549 cells. Compound 11 showed potent cytotoxicity with an IC50 value of 0.16 µM against HL-60 cells and induced apoptotic cell death via a mitochondrion-independent pathway.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos Fitogénicos , Colestanos , Glucósidos , Leucemia Promielocítica Aguda/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Ornithogalum/química , Células A549 , Adenocarcinoma/metabolismo , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/farmacología , Colestanos/química , Colestanos/farmacología , Glucósidos/química , Glucósidos/farmacología , Células HL-60 , Humanos , Leucemia Promielocítica Aguda/metabolismo , Neoplasias Pulmonares/metabolismo
20.
Phytochemistry ; 136: 125-132, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28139298

RESUMEN

Phytochemical investigation of the tubers of Ophiopogon japonicus led to the isolation of five previously undescribed steroidal saponins, ophiojaponins A-E, together with twelve known ones. The structures of these isolated compounds were elucidated by detailed spectroscopic analyses and chemical methods. Ophiojaponins A-C are rare naturally occurring C29 steroidal glycosides possessing a homo-cholestane skeleton with an aromatized ring E. Ruscogenin 1-O-α-L-rhamnopyranosyl-(1 â†’ 2)-4-O-sulfo-ß-D-fucopyranosido-3-O-ß-D-glucopyranoside was isolated as single component and its full spectroscopic data was reported for the first time. The isolated steroidal saponins were evaluated for their cytotoxicities against two human tumor cell lines MG-63 and SNU387. Among them, five known spirostane-type glycosides showed cytotoxic activity against both MG-63 and SNU387 cell lines with IC50 values ranging from 0.76 to 27.0 µM.


Asunto(s)
Antineoplásicos Fitogénicos/aislamiento & purificación , Colestanos/aislamiento & purificación , Medicamentos Herbarios Chinos/aislamiento & purificación , Ophiopogon/química , Tubérculos de la Planta/química , Saponinas/aislamiento & purificación , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/farmacología , Colestanos/química , Colestanos/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Medicamentos Herbarios Chinos/química , Medicamentos Herbarios Chinos/farmacología , Estructura Molecular , Saponinas/química , Saponinas/farmacología , Estereoisomerismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...