Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cardiovasc Toxicol ; 23(3-4): 121-131, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36809482

RESUMEN

With the development and approval of new proteasome inhibitors, proteasome inhibition is increasingly recognized in cancer therapy. Besides successful anti-cancer effects in hematological cancers, side effects such as cardiotoxicity are limiting effective treatment. In this study, we used a cardiomyocyte model to investigate the molecular cardiotoxic mechanisms of carfilzomib (CFZ) and ixazomib (IXZ) alone or in combination with the immunomodulatory drug dexamethasone (DEX) which is frequently used in combination therapies in the clinic. According to our findings, CFZ showed a higher cytotoxic effect at lower concentrations than IXZ. DEX combination attenuated the cytotoxicity for both proteasome inhibitors. All drug treatments caused a marked increase in K48 ubiquitination. Both CFZ and IXZ caused an upregulation in cellular and endoplasmic reticulum stress protein (HSP90, HSP70, GRP94, and GRP78) levels and DEX combination attenuated the increased stress protein levels. Importantly, IXZ and IXZ-DEX treatments caused upregulation of mitochondria fission and fusion gene expression levels higher than caused by CFZ and CFZ-DEX combination. The IXZ-DEX combination reduced the levels of OXPHOS proteins (Complex II-V) more than the CFZ-DEX combination. Reduced mitochondrial membrane potential and ATP production were detected with all drug treatments in cardiomyocytes. Our findings suggest that the cardiotoxic effect of proteasome inhibitors may be due to their class effect and stress response and mitochondrial dysfunction may be involved in the cardiotoxicity process.


Asunto(s)
Antineoplásicos , Inhibidores de Proteasoma , Humanos , Inhibidores de Proteasoma/toxicidad , Cardiotoxicidad , Antineoplásicos/farmacología , Dexametasona/toxicidad , Mitocondrias , Línea Celular Tumoral
2.
Toxicol Appl Pharmacol ; 430: 115723, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34520793

RESUMEN

Pyrethroids are one of the most commonly used classes of synthetic pesticides in the world. Recent laboratory and epidemiological evidence suggested that pyrethroids have potential adverse effects in the mammalian brain; however, the underlying mechanisms of the neurotoxic effects of pyrethroids have not been fully elucidated. In the present study, we investigated the mechanisms of effects of a type II pyrethroid deltamethrin (DM) in a neuronal cell model focusing on the proteolytic function, including autophagy and the ubiquitin-proteasome system. We confirmed that a micromolar concentration of DM dose-dependently decreased the cell viability and induced apoptotic cell death. Our results showed that DM enhanced autophagy in association with an accumulation of autophagosomes and increase in the levels of autophagy markers LC3-II/LC3-I ratio and p62 which were much elevated in the presence of lysosomal inhibitors bafilomycin A1 and chloroquine. We also found that DM caused a dysfunction of mitochondria with a decrease of mitochondrial membrane potential and mitochondrial DNA copy number as well as colocalization with autophagosomes. Moreover, a decrease in the activities of three major proteasomal enzymes and an accumulation of ubiquitinated proteins were observed by the exposure to DM. Transcriptome analysis revealed that up-regulated genes supported the activation of autophagy with induction of cellular stress responses including oxidative stress and endoplasmic reticulum stress, while down-regulated genes related to the cell cycle and DNA replication. These findings provide novel insights into the neurotoxicity of DM which underlie the imbalance in proteolytic function caused by mitophagy activation and proteasome inhibition.


Asunto(s)
Insecticidas/toxicidad , Mitocondrias/efectos de los fármacos , Mitofagia/efectos de los fármacos , Neuronas/efectos de los fármacos , Síndromes de Neurotoxicidad/etiología , Nitrilos/toxicidad , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/toxicidad , Piretrinas/toxicidad , Animales , Apoptosis/efectos de los fármacos , Proteínas Relacionadas con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Estrés del Retículo Endoplásmico/efectos de los fármacos , Regulación de la Expresión Génica , Ratones , Mitocondrias/enzimología , Mitocondrias/genética , Mitocondrias/patología , Neuronas/enzimología , Neuronas/patología , Síndromes de Neurotoxicidad/enzimología , Síndromes de Neurotoxicidad/genética , Síndromes de Neurotoxicidad/patología , Estrés Oxidativo/efectos de los fármacos , Proteolisis , Transcriptoma
3.
Toxicol Appl Pharmacol ; 429: 115695, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34419493

RESUMEN

M3258 is the first selective inhibitor of the immunoproteasome subunit LMP7 (Large multifunctional protease 7) in early clinical development with the potential to improve therapeutic utility in patients of multiple myeloma (MM) or other hematological malignancies. Safety pharmacology studies with M3258 did not reveal any functional impairments of the cardiovascular system in several in vitro tests employing human cardiomyocytes and cardiac ion channels (including hERG), guinea pig heart refractory period and force contraction, and rat aortic contraction as well as in cardiovascular function tests in dogs. Following single dose M3258 administration to rats, no changes were observed on respiratory function by using whole body plethysmography, nor did it change (neuro)behavioral parameters in a battery of tests. Based on pivotal 4-week toxicity studies with daily oral dosing of M3258, the identified key target organs of toxicity were limited to the lympho-hematopoietic system in rats and dogs, and to the intestine with its local lymphoid tissues in dogs only. Importantly, the stomach, nervous system, heart, lungs, and kidneys, that may be part of clinically relevant toxicities as reported for pan-proteasome inhibitors, were spared with M3258. Therefore, it is anticipated that by targeting highly selective and potent inhibition of LMP7, the resulting favorable safety profile of M3258 together with the maintained potent anti-tumor activity as previously reported in mouse MM xenograft models, may translate into an improved benefit-risk profile in MM patients.


Asunto(s)
Ácidos Borónicos/toxicidad , Furanos/toxicidad , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/toxicidad , Administración Oral , Animales , Ácidos Borónicos/administración & dosificación , Células Cultivadas , Perros , Femenino , Furanos/administración & dosificación , Cobayas , Sistema Hematopoyético/efectos de los fármacos , Sistema Hematopoyético/patología , Humanos , Intestinos/efectos de los fármacos , Intestinos/patología , Sistema Linfático/efectos de los fármacos , Sistema Linfático/patología , Masculino , Inhibidores de Proteasoma/administración & dosificación , Ratas Wistar , Medición de Riesgo , Especificidad de la Especie , Pruebas de Toxicidad
4.
Sci Rep ; 11(1): 10523, 2021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-34006972

RESUMEN

Proteasome inhibitors (PIs) represent the gold standard in the treatment of multiple myeloma. Among PIs, Bortezomib (BTZ) is frequently used as first line therapy, but peripheral neuropathy (PN), occurring approximately in 50% of patients, impairs their life, representing a dose-limiting toxicity. Carfilzomib (CFZ), a second-generation PI, induces a significantly less severe PN. We investigated possible BTZ and CFZ off-targets able to explain their different neurotoxicity profiles. In order to identify the possible PIs off-targets we used the SPILLO-PBSS software that performs a structure-based in silico screening on a proteome-wide scale. Among the top-ranked off-targets of BTZ identified by SPILLO-PBSS we focused on tubulin which, by contrast, did not turn out to be an off-target of CFZ. We tested the hypothesis that the direct interaction between BTZ and microtubules would inhibit the tubulin alfa GTPase activity, thus reducing the microtubule catastrophe and consequently furthering the microtubules polymerization. This hypothesis was validated in a cell-free model, since BTZ (but not CFZ) reduces the concentration of the free phosphate released during GTP hydrolysis. Moreover, NMR binding studies clearly demonstrated that BTZ, unlike CFZ, is able to interact with both tubulin dimers and polymerized form. Our data suggest that different BTZ and CFZ neurotoxicity profiles are independent from their proteasome inhibition, as demonstrated in adult mice dorsal root ganglia primary sensory neurons, and, first, we demonstrate, in a cell free model, that BTZ is able to directly bind and perturb microtubules.


Asunto(s)
Antineoplásicos/toxicidad , Bortezomib/toxicidad , Oligopéptidos/toxicidad , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Inhibidores de Proteasoma/toxicidad , Tubulina (Proteína)/metabolismo , Animales , Biopolímeros/metabolismo , Línea Celular , Simulación por Computador , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Humanos , Ratones , Neuronas/efectos de los fármacos , Unión Proteica
5.
Aging (Albany NY) ; 12(22): 22949-22974, 2020 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-33203800

RESUMEN

Proteasome inhibition demonstrates highly effective impact on multiple myeloma (MM) treatment. Here, we aimed to examine anti-tumor efficiency and underlying mechanisms of a novel well tolerated orally applicable proteasome inhibitor NNU546 and its hydrolyzed pharmacologically active form NNU219. NNU219 showed more selective inhibition to proteasome catalytic subunits and less off-target effect than bortezomib ex vivo. Moreover, intravenous and oral administration of either NNU219 or NNU546 led to more sustained pharmacodynamic inhibitions of proteasome activities compared with bortezomib. Importantly, NNU219 exhibited potential anti-MM activity in both MM cell lines and primary samples in vitro. The anti-MM activity of NNU219 was associated with induction of G2/M-phase arrest and apoptosis via activation of the caspase cascade and endoplasmic reticulum stress response. Significant growth-inhibitory effects of NNU219 and NNU546 were observed in 3 different human MM xenograft mouse models. Furthermore, such observation was even found in the presence of a bone marrow microenvironment. Taken together, these findings provided the basis for clinical trial of NNU546 to determine its potential as a candidate for MM treatment.


Asunto(s)
Mieloma Múltiple/tratamiento farmacológico , Inhibidores de Proteasoma/administración & dosificación , Administración Intravenosa , Administración Oral , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Disponibilidad Biológica , Bortezomib/administración & dosificación , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Femenino , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Humanos , Masculino , Ratones , Mieloma Múltiple/enzimología , Mieloma Múltiple/patología , Inhibidores de Proteasoma/farmacocinética , Inhibidores de Proteasoma/toxicidad , Ratas , Transducción de Señal , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Med Chem ; 63(9): 4701-4715, 2020 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-32267687

RESUMEN

While proteasome inhibitors such as bortezomib showed satisfactory clinical benefits in the initial treatment of multiple myeloma (MM), drug resistance and relapse are unavoidable. Recent studies suggested inhibition of histone deacetylases (HDACs) restored sensitivity of bortezomib-resistant MM. Hence, we designed dual inhibitors targeting both HDACs and proteasomes to address the resistance of bortezomib. The most potent inhibitors, ZY-2 and ZY-13 showed excellent inhibition against proteasome and good selectivity against HDACs. In particular, ZY-2 not only exhibited good antiproliferative activities on the MM cell lines RPMI-8226, U266, and KM3 (IC50 values of 6.66, 4.31, and 10.1 nM, respectively) but also showed more potent antiproliferative activities against the bortezomib-resistant MM cell line KM3/BTZ compared with bortezomib (IC50 values of 8.98 vs. 226 nM, P < 0.01) and even better than the combination of the HDAC inhibitor MS-275 and bortezomib (1:1) (IC50 values of 8.98 vs. 98.0 nM, P < 0.01).


Asunto(s)
Antineoplásicos/farmacología , Ácidos Borónicos/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Péptidos/farmacología , Inhibidores de Proteasoma/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/metabolismo , Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Ácidos Borónicos/síntesis química , Ácidos Borónicos/metabolismo , Ácidos Borónicos/toxicidad , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Descubrimiento de Drogas , Ensayos de Selección de Medicamentos Antitumorales , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Histona Desacetilasa 1/química , Histona Desacetilasa 1/metabolismo , Inhibidores de Histona Desacetilasas/síntesis química , Inhibidores de Histona Desacetilasas/metabolismo , Inhibidores de Histona Desacetilasas/toxicidad , Humanos , Simulación del Acoplamiento Molecular , Mieloma Múltiple/tratamiento farmacológico , Péptidos/síntesis química , Péptidos/metabolismo , Péptidos/toxicidad , Complejo de la Endopetidasa Proteasomal/química , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/síntesis química , Inhibidores de Proteasoma/metabolismo , Inhibidores de Proteasoma/toxicidad , Unión Proteica , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Puntos de Control de la Fase S del Ciclo Celular/efectos de los fármacos
7.
ACS Chem Biol ; 15(2): 554-561, 2020 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-31877015

RESUMEN

A considerable number of essential cellular proteins have no catalytic activity and serve instead as structural components to aid in assembling protein complexes. For example, the assembly and function of the 26S proteasome, the major enzymatic complex necessary for ubiquitin-dependent protein degradation, require a number of essential protein contacts to associate the 19S regulatory particle with the 20S core particle. Previously, small molecule inhibitors of the active sites of the 20S core particle have been developed, but the activity of the 26S proteasome could also be altered via the disruption of its assembly. We were interested in discovering a small molecule binder of Rpn-6, as it is among several essential proteins that facilitate 26S assembly, which could be used to further our understanding of the association of the 19S regulatory particle with the 20S core particle. Additionally, we were interested in whether a small molecule-Rpn-6 interaction could potentially be cytotoxic to cancer cells that rely heavily on proteasome activity for survival. A workflow for utilizing a one-bead, one-compound library and a thermal shift assay was developed to discover such a molecule. TXS-8, our lead hit, was discovered to have a low micromolar binding affinity for Rpn-6 as well as very limited binding to other proteins. The cytotoxicity of TXS-8 was evaluated in several cell lines, revealing increased cytotoxicity to hematological cancers. Discovery of this peptoid binder of Rpn-6 provides the initial evidence that Rpn-6 could be a druggable target to affect protein degradation and serves as a primary scaffold from which to design more potent binders. We suspect that Rpn-6 could have additional essential roles beyond that of a molecular clamp of the proteasome to help hematological cancer cells survive and that TXS-8 can serve as a useful tool for further elucidating its roles.


Asunto(s)
Peptidomiméticos/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología , Subunidades de Proteína/metabolismo , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/toxicidad , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Células HEK293 , Humanos , Ligandos , Biblioteca de Péptidos , Peptidomiméticos/metabolismo , Peptidomiméticos/toxicidad , Inhibidores de Proteasoma/metabolismo , Inhibidores de Proteasoma/toxicidad , Unión Proteica
8.
Clin Sci (Lond) ; 133(16): 1827-1844, 2019 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-31409729

RESUMEN

Although proteasome inhibitors (PIs) are modern targeted anticancer drugs, they have been associated with a certain risk of cardiotoxicity and heart failure (HF). Recently, PIs have been combined with anthracyclines (ANTs) to further boost their anticancer efficacy. However, this raised concerns regarding cardiac safety, which were further supported by several in vitro studies on immature cardiomyocytes. In the present study, we investigated the toxicity of clinically used PIs alone (bortezomib (BTZ), carfilzomib (CFZ)) as well as their combinations with an ANT (daunorubicin (DAU)) in both neonatal and adult ventricular cardiomyocytes (NVCMs and AVCMs) and in a chronic rabbit model of DAU-induced HF. Using NVCMs, we found significant cytotoxicity of both PIs around their maximum plasma concentration (cmax) as well as significant augmentation of DAU cytotoxicity. In AVCMs, BTZ did not induce significant cytotoxicity in therapeutic concentrations, whereas the toxicity of CFZ was significant and more profound. Importantly, neither PI significantly augmented the cardiotoxicity of DAU despite even more profound proteasome-inhibitory activity in AVCMs compared with NVCMs. Furthermore, in young adult rabbits, no significant augmentation of chronic ANT cardiotoxicity was noted with respect to any functional, morphological, biochemical or molecular parameter under study, despite significant inhibition of myocardial proteasome activity. Our experimental data show that combination of PIs with ANTs is not accompanied by an exaggerated risk of cardiotoxicity and HF in young adult animal cardiomyocytes and hearts.


Asunto(s)
Antraciclinas/toxicidad , Antineoplásicos/toxicidad , Cardiotoxicidad/etiología , Inhibidores de Proteasoma/toxicidad , Animales , Antraciclinas/administración & dosificación , Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/toxicidad , Bortezomib/administración & dosificación , Bortezomib/toxicidad , Daunorrubicina/administración & dosificación , Daunorrubicina/toxicidad , Relación Dosis-Respuesta a Droga , Masculino , Miocitos Cardíacos/efectos de los fármacos , Oligopéptidos/administración & dosificación , Oligopéptidos/toxicidad , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/administración & dosificación , Conejos , Ratas , Ratas Wistar
9.
Toxicology ; 413: 33-39, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30552955

RESUMEN

Bortezomib, a first-line agent for treatment of multiple myeloma, exhibits anticancer activity through proteasome inhibition. However, bortezomib-induced peripheral neuropathy (BIPN) is one of the most serious side effects. Since decreased proteasomal degradation of Cav3.2 T-type calcium channels in the primary afferents is involved in persistent pain, we investigated whether BIPN involves increased protein levels of Cav3.2 in mice. Six repeated i.p. administrations of bortezomib for 12 days developed persistent mechanical allodynia. Systemic administration of novel T-type calcium channel blockers, (2R/S)-6-prenylnaringenin and KTt-45, and of TTA-A2, the well-known blocker, reversed the BIPN. Ascorbic acid, known to block Cav3.2, but not Cav3.1 or 3.3, and silencing of Cav3.2 gene also suppressed BIPN. Protein levels of Cav3.2 in the dorsal root ganglion (DRG) at L4-L6 levels increased throughout days 1-21 after the onset of bortezomib treatment. Protein levels of USP5, a deubiquitinating enzyme that specifically inhibits proteasomal degradation of Cav3.2, increased in DRG on days 3-21, but not day 1, in bortezomib-treated mice. In DRG-derived ND7/23 cells, bortezomib increased protein levels of Cav3.2 and T-channel-dependent currents, as assessed by a patch-clamp method, but did not upregulate expression of Cav3.2 mRNA or USP5 protein. MG-132, another proteasome inhibitor, also increased Cav3.2 protein levels in the cultured cells. Given the previous evidence for USP5 induction following nociceptor excitation, our data suggest that BIPN involves the increased protein levels of Cav3.2 in nociceptors through inhibition of proteasomal degradation of Cav3.2 by bortezomib itself and then by USP5 that is upregulated probably in an activity-dependent manner.


Asunto(s)
Antineoplásicos/toxicidad , Bortezomib/toxicidad , Canales de Calcio Tipo T/biosíntesis , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Enfermedades del Sistema Nervioso Periférico/metabolismo , Inhibidores de Proteasoma/toxicidad , Animales , Canales de Calcio Tipo T/deficiencia , Canales de Calcio Tipo T/genética , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Técnicas de Silenciamiento del Gen/métodos , Masculino , Ratones , Enfermedades del Sistema Nervioso Periférico/genética , Ratas
10.
Sci Rep ; 8(1): 16318, 2018 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-30397214

RESUMEN

The proteasomal system is responsible for the turnover of damaged proteins. Because of its important functions in oncogenesis, inhibiting the proteasomal system is a promising therapeutic approach for cancer treatment. Bortezomib (BTZ) is the first proteasome inhibitor approved by FDA for clinical applications. However neuropathic side effects are dose limiting for BTZ as many other chemotherapeutic agents. Therefore second-generation proteasome inhibitors have been developed including carfilzomib (CFZ). Aim of the present work was investigating the mechanisms of peripheral neuropathy triggered by the proteasome inhibitor BTZ and comparing the pathways affected by BTZ and CFZ, respectively. Neural stem cells, isolated from the cortex of E14 mouse embryos, were treated with BTZ and CFZ and mass spectrometry was used to compare the global protein pool of treated cells. BTZ was shown to cause more severe cytoskeletal damage, which is crucial in neural cell integrity. Excessive protein carbonylation and actin filament destabilization were also detected following BTZ treatment that was lower following CFZ treatment. Our data on cytoskeletal proteins, chaperone system, and protein oxidation may explain the milder neurotoxic effects of CFZ in clinical applications.


Asunto(s)
Bortezomib/toxicidad , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Neurotoxinas/toxicidad , Oligopéptidos/toxicidad , Inhibidores de Proteasoma/toxicidad , Proteómica , Actinas/metabolismo , Línea Celular , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Células-Madre Neurales/citología , Carbonilación Proteica/efectos de los fármacos , Ubiquitinación/efectos de los fármacos
11.
Cardiovasc Toxicol ; 18(6): 557-568, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29951728

RESUMEN

The proteasome inhibitors bortezomib, carfilzomib, and ixazomib, which are used in the treatment of multiple myeloma have greatly improved response rates. Several other proteasome inhibitors, including delanzomib and oprozomib, are in clinical trials. Carfilzomib and oprozomib are epoxyketones that form an irreversible bond with the 20S proteasome, whereas bortezomib, ixazomib, and delanzomib are boronic acids that form slowly reversible adducts. Several of the proteasome inhibitors have been shown to exhibit specific cardiac toxicities. A primary neonatal rat myocyte model was used to study the relative myocyte-damaging effects of five proteasome inhibitors with a view to identifying potential class differences and the effect of inhibitor binding kinetics. Bortezomib was shown to induce the most myocyte damage followed by delanzomib, ixazomib, oprozomib, and carfilzomib. The sensitivity of myocytes to proteasome inhibitors, which contain high levels of chymotrypsin-like proteasomal activity, may be due to inhibition of proteasomal-dependent ongoing sarcomeric protein turnover. All inhibitors inhibited the chymotrypsin-like proteasomal activity of myocyte lysate in the low nanomolar concentration range and exhibited time-dependent inhibition kinetics characteristic of slow-binding inhibitors. Progress curve analysis of the inhibitor concentration dependence of the slow-binding kinetics was used to measure second-order "on" rate constants for binding. The second-order rate constants varied by 90-fold, with ixazomib reacting the fastest, and oprozomib the slowest. As a group, the boronic acid drugs were more damaging to myocytes than the epoxyketone drugs. Overall, inhibitor-induced myocyte damage was positively, but not significantly, correlated with their second-order rate constants.


Asunto(s)
Ácidos Borónicos/toxicidad , Compuestos Epoxi/toxicidad , Cetonas/toxicidad , Miocitos Cardíacos/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Inhibidores de Proteasoma/toxicidad , Animales , Animales Recién Nacidos , Compuestos de Boro/toxicidad , Bortezomib/toxicidad , Cardiotoxicidad , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Glicina/análogos & derivados , Glicina/toxicidad , Humanos , Células K562 , Cinética , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Oligopéptidos/toxicidad , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Ratas Sprague-Dawley , Treonina/análogos & derivados , Treonina/toxicidad
12.
Chem Res Toxicol ; 31(5): 380-387, 2018 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-29658272

RESUMEN

Activation of the nuclear factor erythroid 2-related factor 2 (Nrf2) antioxidant response signaling pathway is a major mechanism for the cellular defense against oxidative stress. Arsenite, a widespread contaminant in drinking water, is known to induce oxidative stress and activate the Nrf2-dependent signaling pathway through the stabilization of the Nrf2 protein by inhibiting its ubiquitination via the Cul3-Rbx1-Keap1 (cullin 3, RING-box 1, and Kelch-like ECH-associated protein 1) E3 ubiquitin ligase, and its degradation by the 26S proteasome, though the underlying mechanism, remains elusive. In the present study, we demonstrated that arsenite could bind to the RING finger domain of Rbx1 in vitro and in cells, which led to the suppression of Cul3-Rbx1 E3 ubiquitin ligase activity, thereby impairing the Nrf2 ubiquitination and activating the Nrf2-induced antioxidant signaling pathway. Our finding provided novel insight into arsenic toxicity by uncovering a distinct mechanism accounting for arsenite-induced Nrf2 activation.


Asunto(s)
Arsenitos/farmacología , Arsenitos/toxicidad , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología , Dominios RING Finger/efectos de los fármacos , Arsenitos/química , Células Cultivadas , Células HEK293 , Humanos , Microscopía Fluorescente , Inhibidores de Proteasoma/toxicidad , Ubiquitinación/efectos de los fármacos
13.
J Nutr Biochem ; 51: 69-79, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29096318

RESUMEN

Proteasomes are known to degrade proteins involved in various processes like metabolism, signal transduction, cell-cycle regulation, inflammation, and apoptosis. Evidence showed that protein degradation has a strong influence on developing neurons as well as synaptic plasticity. Here, we have shown that sulforaphane (SFN) could prevent the deleterious effects of postnatal proteasomal inhibition on spatial reference and working memory of adult mice. One day old Balb/c mice received intracerebroventricular injections of MG132 and SFN. Sham received an equal volume of aCSF. We observed that SFN pre-administration could attenuate MG132 mediated decrease in proteasome and calpain activities. In vitro findings revealed that SFN could induce proteasomal activity by enhancing the expression of catalytic subunit-ß5. SFN pre-administration prevented the hippocampus based spatial memory impairments during adulthood, mediated by postnatal MG132 exposure. Histological examination showed deleterious effects of MG132 on pyramidal neurons and granule cell neurons in DG and CA3 sub-regions respectively. Furthermore, SFN pre-administration has shown to attenuate the effect of MG132 on proteasome subunit-ß5 expression and also induce the Nrf2 nuclear translocation. In addition, SFN pre-administered mice have also shown to induce expression of pCaMKII, pCreb, and mature/pro-Bdnf, molecules which play a crucial role in spatial learning and memory consolidation. Our findings have shown that proteasomes play an important role in hippocampal synaptic plasticity during the early postnatal period and SFN pre-administration could enhance the proteasomal activity as well as improve spatial learning and memory consolidation.


Asunto(s)
Hipocampo/efectos de los fármacos , Isotiocianatos/uso terapéutico , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Síndromes de Neurotoxicidad/prevención & control , Inhibidores de Proteasoma/toxicidad , Aprendizaje Espacial/efectos de los fármacos , Animales , Animales Recién Nacidos , Región CA3 Hipocampal/efectos de los fármacos , Región CA3 Hipocampal/metabolismo , Región CA3 Hipocampal/patología , Línea Celular Tumoral , Giro Dentado/efectos de los fármacos , Giro Dentado/metabolismo , Giro Dentado/patología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Inyecciones Intraventriculares , Isotiocianatos/administración & dosificación , Leupeptinas/administración & dosificación , Leupeptinas/toxicidad , Trastornos de la Memoria/etiología , Trastornos de la Memoria/prevención & control , Ratones Endogámicos BALB C , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Plasticidad Neuronal/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/farmacología , Síndromes de Neurotoxicidad/metabolismo , Síndromes de Neurotoxicidad/patología , Síndromes de Neurotoxicidad/fisiopatología , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/administración & dosificación , Células Piramidales/efectos de los fármacos , Células Piramidales/metabolismo , Células Piramidales/patología , Sulfóxidos
14.
Neuroscience ; 367: 47-59, 2017 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-29080716

RESUMEN

Ubiquitin-proteasome system (UPS) has emerged as major molecular mechanism which modulates synaptic plasticity. However, very little is known about what happens if this system fails during postnatal brain development. In the present study, MG132 was administered intracerebroventricularly in BALB/c mice pups at postnatal day one (P1), a very crucial period for synaptogenesis. Both 20S proteasome and calpain activities were found to be reduced in the mid brain of MG132-administered pups after 24 h. Mice (P40) which received MG132 on P1 were subjected to Morris water maze (MWM) training. Analysis showed spatial learning and memory of MG132 mice was significantly impaired when compared to age-matched controls. Hematoxylin and eosin as well as Cresyl Violet staining revealed substantial loss of cellular connections, distorted architecture and increased pyknosis in hippocampal CA1 and CA3 regions of MG132 mice. Immunohistochemical analysis of MG132 mice showed increased accumulation of intracellular amyloid-ß in hippocampal cells when compared to control. Moreover, double immunostaining revealed increased expression of amyloid precursor protein C-terminal fragments (APP-CTFß) without affecting ß-secretase expression in MG132 mice. Real-Time PCR analyses showed significant increase in hippocampal expression of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit glutamate A1 (GluA1), but no change in the brain-derived neurotrophic factor (Bdnf) expression in MG132 mice. Western blot analyses showed decreased levels of pThr286-CaMKIIα:CaMKIIα and pSer133-CREB:CREB ratio but increased pro:mature BDNF ratio in the hippocampus of MG132 mice. Taken together, postnatal proteasome inhibition could lead to accumulation of intracellular amyloid-ß protein aggregates, which mediate hippocampus-dependent spatial memory impairments in adult mice.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Hipocampo/metabolismo , Discapacidades para el Aprendizaje/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Aprendizaje Espacial/fisiología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Animales Recién Nacidos , Ácido Aspártico Endopeptidasas/metabolismo , Modelos Animales de Enfermedad , Factor 2 Eucariótico de Iniciación/genética , Factor 2 Eucariótico de Iniciación/metabolismo , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/crecimiento & desarrollo , Discapacidades para el Aprendizaje/inducido químicamente , Leupeptinas/toxicidad , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Inhibidores de Proteasoma/toxicidad , Agregación Patológica de Proteínas/inducido químicamente , Receptores AMPA/genética , Receptores AMPA/metabolismo , Aprendizaje Espacial/efectos de los fármacos
15.
J Steroid Biochem Mol Biol ; 171: 178-186, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28300624

RESUMEN

Steroidal alkaloids are a class of natural products that occur in several species of the Solanaceae family. In the case of the tomato plant (Lycopersicon esculentum Mill.), tomatine and its aglycone, tomatidine, are the most representative molecules. These steroidal alkaloids have already shown several potentially useful biological activities, from anticancer to anti-inflammatory or antibacterial. In this work, the toxicity of these molecules in neuronal cells, namely in the neuroblastoma cell line SH-SY5Y, was assessed, emphasis being given to the cellular mechanisms underlying the effects observed. The results show that tomatine/tomatidine-induced cell death is caspase- and RIP1 kinase-independent, as cell death is not prevented by the pan-caspase inhibitor Z-VAD.fmk or by RIP1 inhibitor necrostatin-1. Analysis of Ca2+ levels using the fluorescent probe Fura-2/AM indicates that both tomatine and tomatidine have a marked effect upon Ca2+ homeostasis by increasing cytosolic Ca2+, an event that might be associated with their effect upon the endoplasmic reticulum. We show that the toxicity of these molecules require the PERK/eIF2α branch of the unfolded protein response, but not the IRE1α branch. Given the role of the endoplasmic reticulum in proteostasis, the ability of these molecules to inhibit the proteasome was also evaluated. Tomatine was able to inhibit the chymotrypsin-like catalytic core of purified human 20S proteasome, as shown by its ability to prevent degradation of the fluorogenic substrate Suc-Leu-Leu-Val-Tyr-AMC, thus suggesting that interference with proteostasis can be responsible for the toxicity of these steroidal alkaloids. This study is relevant as it sheds a light regarding the toxicity of molecules present in one of the most consumed plants worldwide.


Asunto(s)
Retículo Endoplásmico/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/antagonistas & inhibidores , Neuronas/efectos de los fármacos , Inhibidores de Proteasoma/toxicidad , Tomatina/análogos & derivados , Tomatina/toxicidad , Respuesta de Proteína Desplegada/efectos de los fármacos , Apoptosis/efectos de los fármacos , Señalización del Calcio/efectos de los fármacos , Inhibidores de Caspasas/farmacología , Caspasas/química , Caspasas/metabolismo , Línea Celular Tumoral , Nucléolo Celular/efectos de los fármacos , Nucléolo Celular/metabolismo , Tamaño del Núcleo Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Retículo Endoplásmico/enzimología , Retículo Endoplásmico/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/metabolismo , Humanos , Solanum lycopersicum/química , Neuronas/citología , Neuronas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Tomatina/antagonistas & inhibidores
16.
Int J Mol Sci ; 18(4)2017 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-28353647

RESUMEN

Wedelolactone is a multi-target natural plant coumestan exhibiting cytotoxicity towards cancer cells. Although several molecular targets of wedelolactone have been recognized, the molecular mechanism of its cytotoxicity has not yet been elucidated. In this study, we show that wedelolactone acts as an inhibitor of chymotrypsin-like, trypsin-like, and caspase-like activities of proteasome in breast cancer cells. The proteasome inhibitory effect of wedelolactone was documented by (i) reduced cleavage of fluorogenic proteasome substrates; (ii) accumulation of polyubiquitinated proteins and proteins with rapid turnover in tumor cells; and (iii) molecular docking of wedelolactone into the active sites of proteasome catalytic subunits. Inhibition of proteasome by wedelolactone was independent on its ability to induce reactive oxygen species production by redox cycling with copper ions, suggesting that wedelolactone acts as copper-independent proteasome inhibitor. We conclude that the cytotoxicity of wedelolactone to breast cancer cells is partially mediated by targeting proteasomal protein degradation pathway. Understanding the structural basis for inhibitory mode of wedelolactone might help to open up new avenues for design of novel compounds efficiently inhibiting cancer cells.


Asunto(s)
Cumarinas/farmacología , Inhibidores de Proteasoma/farmacología , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cobre/metabolismo , Cumarinas/química , Cumarinas/toxicidad , Humanos , Simulación del Acoplamiento Molecular , Complejo de la Endopetidasa Proteasomal/química , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/química , Inhibidores de Proteasoma/toxicidad , Unión Proteica , Proteolisis , Especies Reactivas de Oxígeno/metabolismo , Ubiquitinación
17.
Neurosci Lett ; 636: 83-89, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27818354

RESUMEN

Lewy bodies, the histopathological hallmarks of Parkinson's disease (PD), contain insoluble and aggregated α-synuclein (aSyn) and many other proteins, proposing a role for failure in protein degradation system in the PD pathogenesis. Proteasomal dysfunction has indeed been linked to PD and aSyn oligomers have been shown to inhibit proteasomes and autophagy. Our recent studies have shown that inhibitors of prolyl oligopeptidase (PREP) can prevent the aggregation and enhance the clearance of accumulated aSyn, and therefore, we wanted to study if PREP inhibition can overcome the aSyn aggregation and toxicity induced by lactacystin, a proteasomal inhibitor. The cells overexpressing human A30P or A53T mutated aSyn were incubated with lactacystin and a PREP inhibitor, KYP-2047, for 48h. Theafter, the cells were fractioned, and the effects of lactacystin with/without 1µM KYP-2047 on aSyn aggregation and ubiquitin accumulation, cell viability and on autophagic markers (p62, Beclin1 and LC3BII) were studied. We found that KYP-2047 attenuated lactacystin-induced cell death in mutant aSyn overexpressing cells but not in non-overexpressing control cells. KYP-2047 reduced significantly SDS-insoluble high-molecular-weight aSyn oligomers that were in line with the cell viability results. In addition, significant reduction in protein accumulation marker, p62, was seen in SDS fraction while LC3BII, a marker for autophagosome formation, was increased, indicating to enhanced autophagy. Our results further streghten the possibilities for PREP inhibitors as a potential drug therapy against synucleinopathies and other protein aggregating diseases.


Asunto(s)
Acetilcisteína/análogos & derivados , Prolina/análogos & derivados , Inhibidores de Proteasoma/toxicidad , Serina Endopeptidasas/metabolismo , Inhibidores de Serina Proteinasa/farmacología , alfa-Sinucleína/metabolismo , Acetilcisteína/toxicidad , Autofagia , Línea Celular Tumoral , Supervivencia Celular , Humanos , Mutación , Prolina/farmacología , Prolil Oligopeptidasas , Agregado de Proteínas , alfa-Sinucleína/genética
18.
Angew Chem Int Ed Engl ; 55(19): 5745-8, 2016 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-27037901

RESUMEN

Elevated expression of the immunoproteasome has been associated with autoimmune diseases, inflammatory diseases, and various types of cancer. Selective inhibitors of the immunoproteasome are not only scarce, but also almost entirely restricted to peptide-based compounds. Herein, we describe nonpeptidic reversible inhibitors that selectively block the chymotrypsin-like (ß5i) subunit of the human immunoproteasome in the low micromolar range. The most potent of the reversibly acting compounds were then converted into covalent, irreversible, nonpeptidic inhibitors that retained selectivity for the ß5i subunit. In addition, these inhibitors discriminate between the immunoproteasome and the constitutive proteasome in cell-based assays. Along with their lack of cytotoxicity, these data point to these nonpeptidic compounds being suitable for further investigation as ß5i-selective probes for possible application in noncancer diseases related to the immunoproteasome.


Asunto(s)
Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células HeLa , Humanos , Concentración 50 Inhibidora , Cinética , Simulación del Acoplamiento Molecular , Oligopéptidos/química , Oligopéptidos/metabolismo , Oligopéptidos/toxicidad , Complejo de la Endopetidasa Proteasomal/química , Inhibidores de Proteasoma/química , Inhibidores de Proteasoma/toxicidad , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/metabolismo , Relación Estructura-Actividad
19.
Nature ; 530(7589): 233-6, 2016 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-26863983

RESUMEN

The proteasome is a multi-component protease complex responsible for regulating key processes such as the cell cycle and antigen presentation. Compounds that target the proteasome are potentially valuable tools for the treatment of pathogens that depend on proteasome function for survival and replication. In particular, proteasome inhibitors have been shown to be toxic for the malaria parasite Plasmodium falciparum at all stages of its life cycle. Most compounds that have been tested against the parasite also inhibit the mammalian proteasome, resulting in toxicity that precludes their use as therapeutic agents. Therefore, better definition of the substrate specificity and structural properties of the Plasmodium proteasome could enable the development of compounds with sufficient selectivity to allow their use as anti-malarial agents. To accomplish this goal, here we use a substrate profiling method to uncover differences in the specificities of the human and P. falciparum proteasome. We design inhibitors based on amino-acid preferences specific to the parasite proteasome, and find that they preferentially inhibit the ß2-subunit. We determine the structure of the P. falciparum 20S proteasome bound to the inhibitor using cryo-electron microscopy and single-particle analysis, to a resolution of 3.6 Å. These data reveal the unusually open P. falciparum ß2 active site and provide valuable information about active-site architecture that can be used to further refine inhibitor design. Furthermore, consistent with the recent finding that the proteasome is important for stress pathways associated with resistance of artemisinin family anti-malarials, we observe growth inhibition synergism with low doses of this ß2-selective inhibitor in artemisinin-sensitive and -resistant parasites. Finally, we demonstrate that a parasite-selective inhibitor could be used to attenuate parasite growth in vivo without appreciable toxicity to the host. Thus, the Plasmodium proteasome is a chemically tractable target that could be exploited by next-generation anti-malarial agents.


Asunto(s)
Antimaláricos/química , Antimaláricos/farmacología , Diseño de Fármacos , Plasmodium/efectos de los fármacos , Plasmodium/enzimología , Inhibidores de Proteasoma/química , Inhibidores de Proteasoma/farmacología , Animales , Antimaláricos/efectos adversos , Antimaláricos/toxicidad , Artemisininas/farmacología , Dominio Catalítico , Microscopía por Crioelectrón , Relación Dosis-Respuesta a Droga , Resistencia a Medicamentos , Sinergismo Farmacológico , Activación Enzimática , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Plasmodium/crecimiento & desarrollo , Plasmodium chabaudi/efectos de los fármacos , Plasmodium chabaudi/enzimología , Plasmodium chabaudi/fisiología , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/enzimología , Plasmodium falciparum/crecimiento & desarrollo , Complejo de la Endopetidasa Proteasomal/química , Complejo de la Endopetidasa Proteasomal/metabolismo , Complejo de la Endopetidasa Proteasomal/ultraestructura , Inhibidores de Proteasoma/efectos adversos , Inhibidores de Proteasoma/toxicidad , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Especificidad de la Especie , Especificidad por Sustrato/efectos de los fármacos
20.
J Oncol Pharm Pract ; 22(5): 720-4, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26044587

RESUMEN

INTRODUCTION: Carfilzomib (Kyprolis™) is a second-generation proteasome inhibitor for the treatment of relapsing multiple myeloma (MM). In 2012, carfilzomib was approved by Food and Drug Administration for the treatment of patients with relapsed MM who had received at least two prior therapies. We present a case of fatal pulmonary toxicity presumed secondary to carfilzomib. CLINICAL PRESENTATION: A 61-year-old male was initially diagnosed with MM in 2003 for which he received multiple treatments. In 2013, he was started on carfilzomib for relapsing MM. After 24 h, the patient developed an acute respiratory distress syndrome for which he needed mechanical ventilation. One week later, patient developed diffuse alveolar hemorrhage and despite aggressive supportive care, the patient died after three weeks. DISCUSSION: The temporal relationship between the first exposure to carfilzomib and development of symptoms, and the exclusion of other possible etiologies, leads us to believe that our patient's lung toxicity is a possible adverse reaction to carfilzomib. To the best of our knowledge, there are no previous reports of deaths due to carfilzomib-related pulmonary toxicity.


Asunto(s)
Antineoplásicos/toxicidad , Oligopéptidos/toxicidad , Inhibidores de Proteasoma/toxicidad , Síndrome de Dificultad Respiratoria/inducido químicamente , Antineoplásicos/uso terapéutico , Resultado Fatal , Humanos , Pulmón/efectos de los fármacos , Pulmón/fisiopatología , Masculino , Persona de Mediana Edad , Mieloma Múltiple/tratamiento farmacológico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Oligopéptidos/uso terapéutico , Inhibidores de Proteasoma/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA