Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Biosci ; 14(1): 61, 2024 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-38735971

RESUMEN

Enolase, a multifunctional protein with diverse isoforms, has generally been recognized for its primary roles in glycolysis and gluconeogenesis. The shift in isoform expression from α-enolase to neuron-specific γ-enolase extends beyond its enzymatic role. Enolase is essential for neuronal survival, differentiation, and the maturation of neurons and glial cells in the central nervous system. Neuron-specific γ-enolase is a critical biomarker for neurodegenerative pathologies and neurological conditions, not only indicating disease but also participating in nerve cell formation and neuroprotection and exhibiting neurotrophic-like properties. These properties are precisely regulated by cysteine peptidase cathepsin X and scaffold protein γ1-syntrophin. Our findings suggest that γ-enolase, specifically its C-terminal part, may offer neuroprotective benefits against neurotoxicity seen in Alzheimer's and Parkinson's disease. Furthermore, although the therapeutic potential of γ-enolase seems promising, the effectiveness of enolase inhibitors is under debate. This paper reviews the research on the roles of γ-enolase in the central nervous system, especially in pathophysiological events and the regulation of neurodegenerative diseases.

2.
Acta Pharm Sin B ; 13(5): 2152-2175, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37250172

RESUMEN

We describe the development of quinolylnitrones (QNs) as multifunctional ligands inhibiting cholinesterases (ChEs: acetylcholinesterase and butyrylcholinesterase-hBChE) and monoamine oxidases (hMAO-A/B) for the therapy of neurodegenerative diseases. We identified QN 19, a simple, low molecular weight nitrone, that is readily synthesized from commercially available 8-hydroxyquinoline-2-carbaldehyde. Quinolylnitrone 19 has no typical pharmacophoric element to suggest ChE or MAO inhibition, yet unexpectedly showed potent inhibition of hBChE (IC50 = 1.06 ± 0.31 nmol/L) and hMAO-B (IC50 = 4.46 ± 0.18 µmol/L). The crystal structures of 19 with hBChE and hMAO-B provided the structural basis for potent binding, which was further studied by enzyme kinetics. Compound 19 acted as a free radical scavenger and biometal chelator, crossed the blood-brain barrier, was not cytotoxic, and showed neuroprotective properties in a 6-hydroxydopamine cell model of Parkinson's disease. In addition, in vivo studies showed the anti-amnesic effect of 19 in the scopolamine-induced mouse model of AD without adverse effects on motoric function and coordination. Importantly, chronic treatment of double transgenic APPswe-PS1δE9 mice with 19 reduced amyloid plaque load in the hippocampus and cortex of female mice, underscoring the disease-modifying effect of QN 19.

3.
Eur J Med Chem ; 249: 115116, 2023 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-36689894

RESUMEN

ATP-competitive inhibitors of human DNA topoisomerase II show potential for becoming the successors of topoisomerase II poisons, the clinically successful anticancer drugs. Based on our recent screening hits, we designed, synthesized and biologically evaluated new, improved series of N-phenylpyrrolamide DNA topoisomerase II inhibitors. Six structural classes were prepared to systematically explore the chemical space of N-phenylpyrrolamide based inhibitors. The most potent inhibitor, 47d, had an IC50 value of 0.67 µM against DNA topoisomerase IIα. Compound 53b showed exceptional activity on cancer cell lines with IC50 values of 130 nM against HepG2 and 140 nM against MCF-7 cancer cell lines. The reported compounds have no structurally similarity to published structures, they are metabolically stable, have reasonable solubility and thus can serve as promising leads in the development of anticancer ATP-competitive inhibitors of human DNA topoisomerase IIα.


Asunto(s)
Antineoplásicos , Humanos , Antineoplásicos/química , Inhibidores de Topoisomerasa II/química , ADN-Topoisomerasas de Tipo II/metabolismo , Adenosina Trifosfato/metabolismo , Antígenos de Neoplasias/metabolismo
4.
Eur J Med Chem ; 247: 115048, 2023 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-36586299

RESUMEN

Alongside reversible butyrylcholinesterase inhibitors, a plethora of covalent butyrylcholinesterase inhibitors have been reported in the literature, typically pseudo-irreversible carbamates. For these latter, however, most cases lack full confirmation of their covalent mode of action. Additionally, the available reports regarding the structure-activity relationships of the O-arylcarbamate warhead are incomplete. Therefore, a follow-up on a series of pseudo-irreversible covalent carbamate human butyrylcholinesterase inhibitors and the structure-activity relationships of the N-dialkyl O-arylcarbamate warhead are presented in this study. The covalent mechanism of binding was tested by IC50 time-dependency profiles, and sequentially and increasingly confirmed by kinetic analysis, whole protein LC-MS, and crystallographic analysis. Computational studies provided valuable insights into steric constraints and identified problematic, bulky carbamate warheads that cannot reach and carbamoylate the catalytic Ser198. Quantum mechanical calculations provided further evidence that steric effects appear to be a key factor in determining the covalent binding behaviour of these carbamate cholinesterase inhibitors and their duration of action. Additionally, the introduction of a clickable terminal alkyne moiety into one of the carbamate N-substituents and in situ derivatisation with azide-containing fluorophore enabled fluorescent labelling of plasma human butyrylcholinesterase. This proof-of-concept study highlights the potential of this novel approach and for these compounds to be further developed as clickable molecular probes for investigating tissue localisation and activity of cholinesterases.


Asunto(s)
Acetilcolinesterasa , Butirilcolinesterasa , Humanos , Butirilcolinesterasa/metabolismo , Cinética , Acetilcolinesterasa/metabolismo , Relación Estructura-Actividad , Inhibidores de la Colinesterasa/farmacología , Inhibidores de la Colinesterasa/química , Carbamatos/farmacología , Carbamatos/química
5.
Biomed Pharmacother ; 149: 112924, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-36068783

RESUMEN

Neurodegenerative diseases are severely debilitating conditions characterized primarily by progressive neuronal loss and impairment of the nervous system. Alzheimer's and Parkinson's diseases are the most common neurodegenerative disorders, and their impact is increasing as average life expectancy increases worldwide. Although the underlying mechanisms of both progressive diseases have been extensively studied, we still lack a comprehensive understanding of the molecular basis of both diseases. Current therapeutic options do not slow the progression of the diseases and only provide symptom relief. Cell models that resemble the characteristics of the disease in question are important in drug discovery projects because they provide information about the therapeutic benefits of drugs under development. Here, we review current in vitro cell models used to study the molecular basis of Alzheimer's and Parkinson's disease focusing on their potential for discovering of disease-modifying therapeutics to combat neurodegenerative diseases. We discuss phenotypic screening as an important approach for identifying novel therapeutic molecules. Advances in the development of cell-based assays for drug discovery are discussed, ranging from simple monoculture cell models to high-throughput three-dimensional cell models. Finally, we critically present the limitations of cell models and the caveats encountered in drug discovery to find effective treatment for neurodegenerative diseases.


Asunto(s)
Enfermedad de Alzheimer , Enfermedades Neurodegenerativas , Enfermedad de Parkinson , Enfermedad de Alzheimer/tratamiento farmacológico , Biología , Descubrimiento de Drogas , Humanos , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedad de Parkinson/tratamiento farmacológico
6.
Int J Mol Sci ; 23(3)2022 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-35163706

RESUMEN

Glioblastoma (GBM) is the most common and deadly primary brain tumor in adults. Understanding GBM pathobiology and discovering novel therapeutic targets are critical to finding efficient treatments. Upregulation of the lysosomal cysteine carboxypeptidase cathepsin X has been linked to immune dysfunction and neurodegenerative diseases, but its role in cancer and particularly in GBM progression in patients is unknown. In this study, cathepsin X expression and activity were found to be upregulated in human GBM tissues compared to low-grade gliomas and nontumor brain tissues. Cathepsin X was localized in GBM cells as well as in tumor-associated macrophages and microglia. Subsequently, potent irreversible (AMS36) and reversible (Z7) selective cathepsin X inhibitors were tested in vitro. Selective cathepsin X inhibitors decreased the viability of patient-derived GBM cells as well as macrophages and microglia that were cultured in conditioned media of GBM cells. We next examined the expression pattern of neuron-specific enzyme γ-enolase, which is the target of cathepsin X. We found that there was a correlation between high proteolytic activity of cathepsin X and C-terminal cleavage of γ-enolase and that cathepsin X and γ-enolase were colocalized in GBM tissues, preferentially in GBM-associated macrophages and microglia. Taken together, our results on patient-derived material suggest that cathepsin X is involved in GBM progression and is a potential target for therapeutic approaches against GBM.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Catepsina Z/metabolismo , Glioblastoma/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Microambiente Tumoral , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Catepsina Z/antagonistas & inhibidores , Catepsina Z/genética , Regulación Neoplásica de la Expresión Génica , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Microglía/efectos de los fármacos , Microglía/metabolismo , Regulación hacia Arriba
7.
FEBS Open Bio ; 12(4): 708-738, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35067006

RESUMEN

Lysosomal peptidases are hydrolytic enzymes capable of digesting waste proteins that are targeted to lysosomes via endocytosis and autophagy. Besides intracellular protein catabolism, they play more specific roles in several other cellular processes and pathologies, either within lysosomes, upon secretion into the cell cytoplasm or extracellular space, or bound to the plasma membrane. In cancer, lysosomal peptidases are generally associated with disease progression, as they participate in crucial processes leading to changes in cell morphology, signaling, migration, and invasion, and finally metastasis. However, they can also enhance the mechanisms resulting in cancer regression, such as apoptosis of tumor cells or antitumor immune responses. Lysosomal peptidases have also been identified as hallmarks of aging and neurodegeneration, playing roles in oxidative stress, mitochondrial dysfunction, abnormal intercellular communication, dysregulated trafficking, and the deposition of protein aggregates in neuronal cells. Furthermore, deficiencies in lysosomal peptidases may result in other pathological states, such as lysosomal storage disease. The aim of this review was to highlight the role of lysosomal peptidases in particular pathological processes of cancer and neurodegeneration and to address the potential of lysosomal peptidases in diagnosing and treating patients.


Asunto(s)
Neoplasias , Péptido Hidrolasas , Apoptosis/fisiología , Humanos , Lisosomas/metabolismo , Procesos Neoplásicos , Péptido Hidrolasas/metabolismo
8.
Bioorg Chem ; 119: 105581, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34990933

RESUMEN

The therapeutic indications for monoamine oxidases A and B (MAO-A and MAO-B) inhibitors that have emerged from biological studies on animal and cellular models of neurological and oncological diseases have focused drug discovery projects upon identifying reversible MAO inhibitors. Screening of our in-house academic compound library identified two hit compounds that inhibit MAO-B with IC50 values in micromolar range. Two series of indole (23 analogues) and 3-(benzyloxy)benzyl)piperazine (16 analogues) MAO-B inhibitors were derived from hits, and screened for their structure-activity relationships. Both series yielded low micromolar selective inhibitors of human MAO-B, namely indole 2 (IC50 = 12.63 ± 1.21 µM) and piperazine 39 (IC50 = 19.25 ± 4.89 µM), which is comparable to selective MAO-B inhibitor isatin (IC50 = 6.10 ± 2.81 µM), yet less potent in comparison to safinamide (IC50 = 0.029 ± 0.002 µM). Selective MAO-B inhibitors 2, 14, 38 and 39 exhibited favourable permeation of the blood-brain barrier and low cytotoxicity in the human neuroblastoma cell line SH-SY5Y.


Asunto(s)
Antineoplásicos/farmacología , Indoles/farmacología , Inhibidores de la Monoaminooxidasa/farmacología , Monoaminooxidasa/metabolismo , Piperazina/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Indoles/síntesis química , Indoles/química , Ratones , Modelos Moleculares , Estructura Molecular , Inhibidores de la Monoaminooxidasa/síntesis química , Inhibidores de la Monoaminooxidasa/química , Nitritos/análisis , Piperazina/síntesis química , Piperazina/química , Relación Estructura-Actividad
9.
Mol Neurobiol ; 59(4): 2258-2276, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35066760

RESUMEN

Microglia are resident macrophages in the central nervous system that are involved in immune responses driven by Toll-like receptors (TLRs). Microglia-mediated inflammation can lead to central nervous system disorders, and more than one TLR might be involved in these pathological processes. The cysteine peptidase cathepsin X has been recognized as a pathogenic factor for inflammation-induced neurodegeneration. Here, we hypothesized that simultaneous TLR3 and TLR4 activation induces synergized microglia responses and that these phenotype changes affect cathepsin X expression and activity. Murine microglia BV2 cells and primary murine microglia were exposed to the TLR3 ligand polyinosinic-polycytidylic acid (poly(I:C)) and the TLR4 ligand lipopolysaccharide (LPS), individually and simultaneously. TLR3 and TLR4 co-activation resulted in increased inflammatory responses compared to individual TLR activation, where poly(I:C) and LPS induced distinct patterns of proinflammatory factors together with different patterns of cathepsin X expression and activity. TLR co-activation decreased intracellular cathepsin X activity and increased cathepsin X localization at the plasma membrane with concomitant increased extracellular cathepsin X protein levels and activity. Inhibition of cathepsin X in BV2 cells by AMS36, cathepsin X inhibitor, significantly reduced the poly(I:C)- and LPS-induced production of proinflammatory cytokines as well as apoptosis. Additionally, inhibiting the TLR3 and TLR4 common signaling pathway, PI3K, with LY294002 reduced the inflammatory responses of the poly(I:C)- and LPS-activated microglia and recovered cathepsin X activity. We here provide evidence that microglial cathepsin X strengthens microglia activation and leads to subsequent inflammation-induced neurodegeneration. As such, cathepsin X represents a therapeutic target for treating neurodegenerative diseases related to excess inflammation.


Asunto(s)
Microglía , Receptor Toll-Like 3 , Animales , Cisteína/metabolismo , Inflamación/metabolismo , Ligandos , Lipopolisacáridos/farmacología , Ratones , Microglía/metabolismo , Poli I-C/efectos adversos , Poli I-C/metabolismo , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 4/metabolismo
10.
Biomolecules ; 11(12)2021 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-34944440

RESUMEN

Neuroinflammation, which is mediated by microglia and astrocytes, is associated with the progression of neurodegenerative diseases. Increasing evidence shows that activated microglia induce the expression and secretion of various lysosomal cathepsins, particularly during the early stage of neuroinflammation. This trigger signaling cascade that aggravate neurodegeneration. To date, most research on neuroinflammation has focused on the role of cysteine cathepsins, the largest cathepsin family. Cysteine cathepsins are primarily responsible for protein degradation in lysosomes; however, they also play a role in regulating a number of other important physiological and pathological processes. This review focuses on the functional roles of cysteine cathepsins in the central nervous system during neuroinflammation, with an emphasis on their roles in the polarization of microglia and neuroinflammation signaling, which in turn causes neuronal death and thus neurodegeneration.


Asunto(s)
Proteasas de Cisteína/metabolismo , Microglía/fisiología , Enfermedades Neuroinflamatorias/fisiopatología , Progresión de la Enfermedad , Regulación de la Expresión Génica , Humanos , Lisosomas/metabolismo , Microglía/metabolismo , Enfermedades Neuroinflamatorias/metabolismo , Proteolisis
11.
Cell Commun Signal ; 19(1): 118, 2021 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-34895236

RESUMEN

BACKGROUND: Neurotrophins can activate multiple signalling pathways in neuronal cells through binding to their cognate receptors, leading to neurotrophic processes such as cell survival and differentiation. γ-Enolase has been shown to have a neurotrophic activity that depends on its translocation towards the plasma membrane by the scaffold protein γ1-syntrophin. The association of γ-enolase with its membrane receptor or other binding partners at the plasma membrane remains unknown. METHODS: In the present study, we used immunoprecipitation and immunofluorescence to show that γ-enolase associates with the intracellular domain of the tropomyosin receptor kinase (Trk) family of tyrosine kinase receptors at the plasma membrane of differentiated SH-SY5Y cells. RESULTS: In differentiated SH-SY5Y cells with reduced expression of γ1-syntrophin, the association of γ-enolase with the Trk receptor was diminished due to impaired translocation of γ-enolase towards the plasma membrane or impaired Trk activity. Treatment of differentiated SH-SY5Y cells with a γ-Eno peptide that mimics γ-enolase neurotrophic activity promoted Trk receptor internalisation and endosomal trafficking, as defined by reduced levels of Trk in clathrin-coated vesicles and increased levels in late endosomes. In this way, γ-enolase triggers Rap1 activation, which is required for neurotrophic activity of γ-enolase. Additionally, the inhibition of Trk kinase activity by K252a revealed that increased SH-SY5Y cell survival and neurite outgrowth mediated by the γ-Eno peptide through activation of signalling cascade depends on Trk kinase activity. CONCLUSIONS: These data therefore establish the Trk receptor as a binding partner of γ-enolase, whereby Trk endosomal trafficking is promoted by γ-Eno peptide to mediate its neurotrophic signalling. Video abstract.


Asunto(s)
Diferenciación Celular , Neuritas , Fosfopiruvato Hidratasa , Receptor trkA , Línea Celular Tumoral , Humanos , Neuritas/fisiología , Fosfopiruvato Hidratasa/metabolismo , Receptor trkA/metabolismo
12.
Int J Mol Sci ; 22(24)2021 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-34948293

RESUMEN

Cathepsin X is a lysosomal peptidase that is involved in tumour progression and represents a potential target for therapeutic interventions. In addition, it regulates important functions of immune cells and is implicated in the modulation of tumour cell-immune cell crosstalk. Selective cathepsin X inhibitors have been proposed as prospective antitumour agents to prevent cancer progression; however, their impact on the antitumour immune response has been overlooked. Previous studies indicate that the migration and adhesion of T cells and dendritic cells are affected by diminished cathepsin X activity. Meanwhile, the influence of cathepsin X inhibition on natural killer (NK) cell function has not yet been explored. Here, we examined the localization patterns of cathepsin X and the role of its inhibitors on the cytotoxicity of cell line NK-92, which is used for adoptive cellular immunotherapy in cancer patients. NK-92 cells depend on lymphocyte function-associated antigen 1 (LFA-1) to form stable immunoconjugates with target cells, providing, in this way, optimal cytotoxicity. Since LFA-1 is a substrate for cathepsin X activity in other types of cells, we hypothesized that cathepsin X could disturb the formation of NK-92 immunoconjugates. Thus, we employed cathepsin X reversible and irreversible inhibitors and evaluated their effects on the NK-92 cell interactions with target cells and on the NK-92 cell cytotoxicity. We show that cathepsin X inhibition does not impair stable conjugate formation or the lytic activity of NK-92 cells. Similarly, the conjugate formation between Jurkat T cells and target cells was not affected by cathepsin X activity. Unlike in previous migration and adhesion studies on T cells, in NK-92 cells cathepsin X was not co-localized with LFA-1 at the plasma membrane but was, rather, redistributed to the cytotoxic granules and secreted during degranulation.


Asunto(s)
Catepsinas/farmacología , Gránulos Citoplasmáticos/efectos de los fármacos , Citotoxicidad Inmunológica/efectos de los fármacos , Células Asesinas Naturales/efectos de los fármacos , Sinapsis/efectos de los fármacos , Antineoplásicos/farmacología , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Humanos , Inmunoterapia Adoptiva/métodos , Células Jurkat , Células K562 , Neoplasias/tratamiento farmacológico , Linfocitos T/efectos de los fármacos
13.
Crit Rev Immunol ; 41(2): 77-99, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34348003

RESUMEN

Natural killer (NK) cells represent critical effectors of anti-tumor immune responses due to their ability to target tumor cells that escape recognition by the adaptive arm of the immune system. NK cell efficacy depends on multiple factors, including their propensity to infiltrate tumors, to reach activation threshold, and to differentiate into mature cytotoxic cells. The tumor microenvironment counteracts protective immunity by delivering anti-inflammatory signals, which stimulate the development of myeloid-derived suppressor cells (MDSC). MDSCs utilize numerous proximity-dependent and independent mechanisms to suppress functions of cytotoxic T lymphocytes and NK cells. Importantly, substantial part of their suppressive activity depends on peptidases. MDSC-derived peptidases incapacitate NK cells by shedding ligands for their activating receptors and by processing key cytokines involved in regulation of immune responses. Moreover, they are needed for sustaining the immunosuppressive loop through promotion of MDSC accumulation, expansion, and enhancement of their survival. Peptidases are at the forefront of cancer progression. However, their disparate roles in immune cells have only recently become appreciated in orchestration of the cancer immune responses. Studies that focused on elucidating the potential of peptidase inhibitors in regulation of the anti-tumor immune responses have led to renewed interest in clinical development of peptidase inhibitors. In parallel, they inspired the development of novel theranostics, that exploit increased activity of peptidases in infiltrating immune cells for targeted drug release or activation of imaging probes.


Asunto(s)
Células Supresoras de Origen Mieloide , Neoplasias , Humanos , Células Asesinas Naturales , Neoplasias/terapia , Péptido Hidrolasas , Microambiente Tumoral
14.
Eur J Med Chem ; 209: 112911, 2021 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-33071056

RESUMEN

Structurally diverse heterotricyclic compounds are recognized as monoamine oxidase (MAO) inhibitors and thus represent an appealing scaffold in development and optimization of novel MAO inhibitors. Herein we explored the chemical space of pyrimido[1,2-b]indazoles as MAO inhibitors by preparing a small library of (hetero)aryl derivatives. An efficient synthetic strategy was developed starting from commercially available 1H-indazol-3-amines, which were converted to various 3-bromoheterotricyclic derivatives and further functionalized via Suzuki-Miyaura coupling reaction. Derivatives 4a-t selectively inhibited human MAO-B isoform in a reversible and competitive manner as confirmed by kinetic experiments and docking studies. Selected derivatives were not cytotoxic to neuroblastoma SH-SY5Y cells. Moreover, analogue 4i protected human neuroblastoma SH-SY5Y cells against 6-hydroxydopamine-induced cell death, which confirms the applicability of the pyrimido[1,2-b]indazoles as potential antiparkinsonian agents.


Asunto(s)
Antiparkinsonianos/síntesis química , Indazoles/síntesis química , Inhibidores de la Monoaminooxidasa/síntesis química , Monoaminooxidasa/metabolismo , Neuroblastoma/tratamiento farmacológico , Fármacos Neuroprotectores/síntesis química , Bibliotecas de Moléculas Pequeñas/síntesis química , Antiparkinsonianos/química , Antiparkinsonianos/farmacología , Bromuros/química , Complejos de Coordinación/química , Humanos , Indazoles/química , Indazoles/farmacocinética , Metales/química , Simulación del Acoplamiento Molecular , Inhibidores de la Monoaminooxidasa/farmacocinética , Fármacos Neuroprotectores/farmacocinética , Unión Proteica , Bibliotecas de Moléculas Pequeñas/farmacocinética , Relación Estructura-Actividad , Tiramina/química
15.
Front Mol Neurosci ; 13: 575453, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33328882

RESUMEN

Neuroinflammation is an important factor in the pathogenesis of neurodegenerative diseases. Microglia-derived lysosomal cathepsins have been increasingly recognized as important inflammatory mediators that trigger signaling pathways that aggravate neuroinflammation. In vitro, a contribution to neuroinflammation processes has been shown for cathepsin X: however, the expression patterns and functional role of cathepsin X in neuroinflammatory brain pathology remain elusive. In this study we analyzed the expression, activity, regional distribution and cellular localization of cathepsin X in the rat brain with neuroinflammation-induced neurodegeneration. The unilateral injection of lipopolysaccharide (LPS) induced a strong upregulation of cathepsin X expression and its activity in the ipsilateral striatum. In addition to the striatum, cathepsin X overexpression was detected in other brain areas such as the cerebral cortex, corpus callosum, subventricular zone and external globus pallidus, whereas the upregulation was mainly restricted to activated microglia and reactive astrocytes. Continuous administration of the cathepsin X inhibitor AMS36 indicated protective effects against LPS-induced striatal degeneration, as seen by the attenuated LPS-mediated dilation of the lateral ventricles and partial decreased extent of striatal lesion. Taken together, our results indicate that cathepsin X plays a role as a pathogenic factor in neuroinflammation-induced neurodegeneration and represents a potential therapeutic target for neurodegenerative diseases associated with neuroinflammation.

16.
PLoS Pathog ; 16(11): e1009013, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33137165

RESUMEN

Over the last 2 decades, several coronaviruses (CoVs) have crossed the species barrier into humans, causing highly prevalent and severe respiratory diseases, often with fatal outcomes. CoVs are a large group of enveloped, single-stranded, positive-sense RNA viruses, which encode large replicase polyproteins that are processed by viral peptidases to generate the nonstructural proteins (Nsps) that mediate viral RNA synthesis. Papain-like peptidases (PLPs) and chymotrypsin-like cysteine 3C-like peptidase are essential for coronaviral replication and represent attractive antiviral drug targets. Furthermore, CoVs utilize the activation of their envelope spike glycoproteins by host cell peptidases to gain entry into cells. CoVs have evolved multiple strategies for spike protein activation, including the utilization of lysosomal cysteine cathepsins. In this review, viral and host peptidases involved in CoV cell entry and replication are discussed in depth, with an emphasis on papain-like cysteine cathepsins. Furthermore, important findings on cysteine peptidase inhibitors with regard to virus attenuation are highlighted as well as the potential of such inhibitors for future treatment strategies for CoV-related diseases.


Asunto(s)
Antivirales/farmacología , Infecciones por Coronavirus/virología , Coronavirus/efectos de los fármacos , Inhibidores de Cisteína Proteinasa/farmacología , Internalización del Virus/efectos de los fármacos , Animales , Infecciones por Coronavirus/tratamiento farmacológico , Humanos , Replicación Viral/efectos de los fármacos
17.
J Enzyme Inhib Med Chem ; 35(1): 1944-1952, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33092411

RESUMEN

Effective therapy of Alzheimer's disease (AD) requires treatment with a combination of drugs that modulate various pathomechanisms contributing to the disease. In our research, we have focused on the development of multi-target-directed ligands - 5-HT6 receptor antagonists and cholinesterase inhibitors - with disease-modifying properties. We have performed extended in vitro (FRET assay) and in cellulo (Escherichia coli model of protein aggregation) studies on their ß-secretase, tau, and amyloid ß aggregation inhibitory activity. Within these multifunctional ligands, we have identified compound 17 with inhibitory potency against tau and amyloid ß aggregation in in cellulo assay of 59% and 56% at 10 µM, respectively, hBACE IC50=4 µM, h5TH6 K i=94 nM, hAChE IC50=26 nM, and eqBuChE IC50=5 nM. This study led to the development of multifunctional ligands with a broad range of biological activities crucial not only for the symptomatic but also for the disease-modifying treatment of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Inhibidores de la Colinesterasa/química , Colinesterasas/metabolismo , Receptores de Serotonina/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/prevención & control , Inhibidores de la Colinesterasa/metabolismo , Diseño de Fármacos , Escherichia coli , Transferencia Resonante de Energía de Fluorescencia , Humanos , Ligandos , Modelos Moleculares , Agregado de Proteínas , Relación Estructura-Actividad
18.
Cancer Immunol Immunother ; 69(9): 1869-1880, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32372139

RESUMEN

Increased proteolytic activity of cysteine cathepsins has long been known to facilitate malignant progression, and it has also been associated with tumor-promoting roles of myeloid-derived suppressor cells (MDSCs). Consequently, cysteine cathepsins have gained much attention as potential targets for cancer therapies. However, cross-talk between tumor cells and MDSCs needs to be taken into account when studying the efficacy of cathepsin inhibitors as anti-cancer agents. Here, we demonstrate the potential of the MDA-MB-231 breast cancer cell line to generate functional MDSCs from CD14+ cells of healthy human donors. During this transition to MDSCs, the overall levels of cysteine cathepsins increased, with the largest responses for cathepsins L and X. We used small-molecule inhibitors of cathepsins L and X (i.e., CLIK-148, Z9, respectively) to investigate their functional impact on tumor cells and immune cells in this co-culture system. Interactions with peripheral blood mononuclear cells reduced MDA-MB-231 cell invasion, while inhibition of cathepsin X activity by Z9 restored invasion. Inhibition of cathepsin L activity using CLIK-148 resulted in significantly increased CD8+ cytotoxicity. Of note, inhibition of cathepsins L and X in separate immune or tumor cells did not promote these functional changes. Together, our findings underlie the importance of tumor cell-immune cell interactions in the evaluation of the anti-cancer potential of cysteine cathepsin inhibitors.


Asunto(s)
Catepsina L/metabolismo , Cisteína/metabolismo , Células Supresoras de Origen Mieloide/metabolismo , Neoplasias/metabolismo , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Humanos , Leucocitos Mononucleares/metabolismo , Invasividad Neoplásica/patología , Neoplasias/patología , Células PC-3
19.
Eur J Med Chem ; 197: 112282, 2020 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-32380361

RESUMEN

Compounds capable of interacting with single or multiple targets involved in Alzheimer's disease (AD) pathogenesis are potential anti-Alzheimer's agents. In our aim to develop new anti-Alzheimer's agents, a series of 36 new N-alkylpiperidine carbamates was designed, synthesized and evaluated for the inhibition of cholinesterases [acetylcholinesterase (AChE) and butyrylcholinesterase (BChE)] and monoamine oxidases [monoamine oxidase A (MAO-A and monoamine oxidase B (MAO-B)]. Four compounds are very promising: multiple AChE (IC50 = 7.31 µM), BChE (IC50 = 0.56 µM) and MAO-B (IC50 = 26.1 µM) inhibitor 10, dual AChE (IC50 = 2.25 µM) and BChE (IC50 = 0.81 µM) inhibitor 22, selective BChE (IC50 = 0.06 µM) inhibitor 13, and selective MAO-B (IC50 = 0.18 µM) inhibitor 16. Results of enzyme kinetics experiments showed that despite the carbamate group in the structure, compounds 10, 13, and 22 are reversible and non-time-dependent inhibitors of AChE and/or BChE. The resolved crystal structure of the complex of BChE with compound 13 confirmed the non-covalent mechanism of inhibition. Additionally, N-propargylpiperidine 16 is an irreversible and time-dependent inhibitor of MAO-B, while N-benzylpiperidine 10 is reversible. Additionally, compounds 10, 13, 16, and 22 should be able to cross the blood-brain barrier and are not cytotoxic to human neuronal-like SH-SY5Y and liver HepG2 cells. Finally, compounds 10 and 16 also prevent amyloid ß1-42 (Aß1-42)-induced neuronal cell death. The neuroprotective effects of compound 16 could be the result of its Aß1-42 anti-aggregation effects.


Asunto(s)
Carbamatos/farmacología , Inhibidores de la Colinesterasa/farmacología , Inhibidores de la Monoaminooxidasa/farmacología , Fármacos Neuroprotectores/farmacología , Piperidinas/farmacología , Acetilcolinesterasa/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/metabolismo , Butirilcolinesterasa/metabolismo , Carbamatos/síntesis química , Carbamatos/toxicidad , Línea Celular Tumoral , Inhibidores de la Colinesterasa/síntesis química , Inhibidores de la Colinesterasa/toxicidad , Diseño de Fármacos , Humanos , Estructura Molecular , Inhibidores de la Monoaminooxidasa/síntesis química , Inhibidores de la Monoaminooxidasa/toxicidad , Fármacos Neuroprotectores/síntesis química , Fármacos Neuroprotectores/toxicidad , Fragmentos de Péptidos/metabolismo , Piperidinas/síntesis química , Piperidinas/toxicidad , Multimerización de Proteína/efectos de los fármacos , Relación Estructura-Actividad
20.
J Med Chem ; 63(3): 1361-1387, 2020 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-31917923

RESUMEN

The resurgence of interest in monoamine oxidases (MAOs) has been fueled by recent correlations of this enzymatic activity with cardiovascular, neurological, and oncological disorders. This has promoted increased research into selective MAO-A and MAO-B inhibitors. Here, we shed light on how selective inhibition of MAO-A and MAO-B can be achieved by geometric isomers of cis- and trans-1-propargyl-4-styrylpiperidines. While the cis isomers are potent human MAO-A inhibitors, the trans analogues selectively target only the MAO-B isoform. The inhibition was studied by kinetic analysis, UV-vis spectrum measurements, and X-ray crystallography. The selective inhibition of the MAO-A and MAO-B isoforms was confirmed ex vivo in mouse brain homogenates, and additional in vivo studies in mice show the therapeutic potential of 1-propargyl-4-styrylpiperidines for central nervous system disorders. This study represents a unique case of stereoselective activity of cis/trans isomers that can discriminate between structurally related enzyme isoforms.


Asunto(s)
Antidepresivos/uso terapéutico , Depresión/tratamiento farmacológico , Inhibidores de la Monoaminooxidasa/uso terapéutico , Piperidinas/uso terapéutico , Estirenos/uso terapéutico , Animales , Antidepresivos/síntesis química , Antidepresivos/metabolismo , Encéfalo , Dominio Catalítico , Humanos , Isoenzimas/antagonistas & inhibidores , Cinética , Masculino , Ratones , Simulación del Acoplamiento Molecular , Estructura Molecular , Monoaminooxidasa/química , Monoaminooxidasa/clasificación , Monoaminooxidasa/metabolismo , Inhibidores de la Monoaminooxidasa/síntesis química , Inhibidores de la Monoaminooxidasa/metabolismo , Piperidinas/síntesis química , Piperidinas/metabolismo , Unión Proteica , Estereoisomerismo , Relación Estructura-Actividad , Estirenos/síntesis química , Estirenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...