Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
1.
ACS Appl Mater Interfaces ; 16(19): 24295-24307, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38697643

RESUMEN

Pyroptosis has garnered increasing attention because of its ability to trigger robust antitumor immunity. Pyroptosis is initiated by the activation of inflammasomes, which are regulated by various organelles. The collaboration among organelles offers several protective mechanisms to prevent activation of the inflammasome, thereby limiting the induction of efficient pyroptosis. Herein, a multiorganelle homeostasis disruptor (denoted BLL) is constructed by encapsulating liposomes and bortezomib (BTZ) within a layered double hydroxide (LDH) nanocage to continuously activate inflammasomes for inducing efficient pyroptosis. In lysosomes, the negatively charged liposomes are released to recruit the NLRP3 inflammasomes through electrostatic interactions. ER stress is induced by BTZ to enhance the activation of the NLRP3 inflammasome. Meanwhile, the BLL nanocage exhibited H+-scavenging ability due to the weak alkalinity of LDH, thus disrupting the homeostasis of the lysosome and alleviating the degradation of the NLRP3 inflammasome by lysosomal-associated autophagy. Our results suggest that the BLL nanocage induces homeostatic imbalance in various organelles and efficient pyroptosis. We hope this work can provide new insights into the design of an efficient pyroptosis inducer by disrupting the homeostatic balance of multiple organelles and promote the development of novel antineoplastic platforms.


Asunto(s)
Homeostasis , Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Piroptosis , Piroptosis/efectos de los fármacos , Inflamasomas/metabolismo , Inflamasomas/efectos de los fármacos , Homeostasis/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Humanos , Ratones , Bortezomib/farmacología , Bortezomib/química , Liposomas/química , Animales , Lisosomas/metabolismo , Lisosomas/efectos de los fármacos , Hidróxidos/química , Hidróxidos/farmacología , Nanoestructuras/química , Nanopartículas/química
2.
Mol Pharm ; 20(1): 524-544, 2023 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-36306447

RESUMEN

Breast cancer leads to the highest mortality among women resulting in a major clinical burden. Multidrug therapy is more efficient in such patients compared to monodrug therapy. Simultaneous combinatorial or co-delivery garnered significant interest in the past years. Caffeic acid (CFA) (a natural polyphenol) has received growing attention because of its anticarcinogenic and antioxidant potential. Bortezomib (BTZ) is a proteasome inhibitor and may be explored for treating breast cancer. Despite its high anticancer activity, the low water solubility and chemical instability restrict its efficacy against solid tumors. In the present study, we designed and investigated a HP-PCL (N-2-hydroxypropylmethacrylamide-polycaprolactone) polymeric micellar (PMCs) system for the simultaneous delivery of BTZ and CFA in the treatment of breast cancer. The designed BTZ+CFA-HP-PCL PMCs were fabricated, optimized, and characterized for size, zeta potential, surface morphology, and in vitro drug release. Developed nanosized (174.6 ± 0.24 nm) PMCs showed enhanced cellular internalization and cell cytotoxicity in both MCF-7 and MDA-MB-231 cells. ROS (reactive oxygen species) levels were highest in BTZ-HP-PCL PMCs, while CFA-HP-PCL PMCs significantly (p < 0.001) scavenged the ROS generated in 2',7'-dichlorofluorescein diacetate (DCFH-DA) assay. The mitochondrial membrane potential (MMP) assay revealed intense and significant green fluorescence in both types of cancer cells when treated with BTZ-HP-PCL PMCs (p < 0.001) indicating apoptosis or cell death. The pharmacokinetic studies revealed that BTZ-HP-PCL PMCs and BTZ+CFA-HP-PCL PMCs exhibited the highest bioavailability, enhanced plasma half-life, decreased volume of distribution, and lower clearance rate than the pure combination of drugs. In the organ biodistribution studies, the combination of BTZ+CFA showed higher distribution in the spleen and the heart. Overall findings of in vitro studies surprisingly resulted in better therapeutic efficiency of BTZ-HP-PCL PMCs than BTZ+CFA-HP-PCL PMCs. However, the in vivo tumor growth inhibition study performed in tumor-induced mice concluded that the tumor growth was inhibited by both BTZ-HP-PCL PMCs and BTZ+CFA-HP-PCL PMCs (p < 0.0001) more efficiently than pure BTZ and the combination (BTZ+CFA), which may be due to the conversion of boronate ester into boronic acid. Henceforth, the combination of BTZ and CFA provides further indications to be explored in the future to support the hypothesis that BTZ may work with polyphenol (CFA) in the acidic environment of the tumor.


Asunto(s)
Antineoplásicos , Inhibidores de Proteasoma , Femenino , Ratones , Animales , Inhibidores de Proteasoma/farmacología , Inhibidores de Proteasoma/uso terapéutico , Micelas , Especies Reactivas de Oxígeno , Distribución Tisular , Quimioterapia Combinada , Leprostáticos/uso terapéutico , Bortezomib/farmacología , Bortezomib/química , Polímeros/química , Línea Celular Tumoral , Antineoplásicos/química
3.
Biomaterials ; 288: 121737, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36031455

RESUMEN

The clinical translation of nanomedicines has been impeded by the unfavorable tumor microenvironment (TME), particularly the tortuous vasculature networks, which significantly influence the transport and distribution of nanomedicines into tumors. In this work, a smart pH-responsive bortezomib (BTZ)-loaded polyhydralazine nanoparticle (PHDZ/BTZ) is presented, which has a great capacity to augment the accumulation of BTZ in tumors by dilating tumor blood vessels via specific release of vasodilator hydralazine (HDZ). The Lewis acid-base coordination effect between the boronic bond of BTZ and amino of HDZ empowered PHDZ/BTZ nanoparticles with great stability and high drug loading contents. Once triggered by the acidic tumor environment, HDZ could be released quickly to remodel TME through tumor vessel dilation, hypoxia attenuation, and lead to an increased intratumoral BTZ accumulation. Additionally, our investigation revealed that this pH-responsive nanoparticle dramatically suppressed tumor growth, inhibited the occurrence of lung metastasis with fewer side effects and induced immunogenic cell death (ICD), thereby eliciting immune activation including massive cytotoxic T lymphocytes (CTLs) infiltration in tumors and efficient serum proinflammatory cytokine secretion compared with free BTZ treatment. Thus, with efficient drug loading capacity and potent immune activation, PHDZ nanoparticles exhibit great potential in the delivery of boronic acid-containing drugs aimed at a wide range of diseases.


Asunto(s)
Antineoplásicos , Nanopartículas , Neoplasias , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Bortezomib/química , Bortezomib/farmacología , Bortezomib/uso terapéutico , Línea Celular Tumoral , Humanos , Concentración de Iones de Hidrógeno , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Microambiente Tumoral
4.
Yakugaku Zasshi ; 142(2): 145-153, 2022.
Artículo en Japonés | MEDLINE | ID: mdl-35110451

RESUMEN

The first medicine containing the boron element, bortezomib, was approved for clinical use just 18 years ago. The boronic acid substructure in bortezomib serves as an electrophilic functionality with high affinity for hydroxy groups, which are frequently found in catalytic sites of proteolytic enzymes, to create reversible covalent bonds with a slow dissociation rate. Today, boronic acid is considered an important molecule in the medicinal chemistry toolbox, which was promoted by the success of bortezomib and pioneering approaches to use boronic acid in the molecular design of serine protease inhibitors in the 1980s. In this review article, we first provide an overview of the development of bortezomib, and then summarize our achievements to construct boronic acid analogs of tyropeptin A, a naturally occurring proteasome inhibitor, with potent in vivo efficacy. Representative stereoselective synthetic methods of α-aminoboronic acid are also showcased.


Asunto(s)
Antineoplásicos/síntesis química , Ácidos Borónicos/química , Bortezomib/síntesis química , Desarrollo de Medicamentos/métodos , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Inhibidores de Serina Proteinasa/síntesis química , Bortezomib/química , Catálisis , Dipéptidos/síntesis química , Dipéptidos/química , Diseño de Fármacos , Inhibidores de Serina Proteinasa/química , Estereoisomerismo
5.
Yakugaku Zasshi ; 142(2): 131-137, 2022.
Artículo en Japonés | MEDLINE | ID: mdl-35110449

RESUMEN

Almost all conventional drug discovery research has been based on hydrocarbon-based frameworks and common chemical elements such as nitrogen, oxygen, sulfur, and the halogens. However, triggered by the approval of bortezomib, a boronic acid-containing pharmaceutical agent, the incorporation of functionalities that are not native in biological systems has been intensively investigated. Several other boron-containing pharmaceuticals have also been marketed. Therefore, the inclusion of various elements is one of the most promising strategies for the development of novel and distinctive drug candidates. In this symposium review, the author focused on the 'elements chemistry' approaches for the structural development of biologically active compounds, particularly those involving silicon and phosphorus. The isosteric exchange of Si and C (Si/C-exchange) is one of the most-investigated forms of substituting elements. We revealed the detailed physicochemical impact of Si/C-exchange, and we proposed several applications of silyl functionalities other than the simple Si/C-exchange. Regarding phosphorus, we recently revealed that the P-B substructure can function as the isostere of C-C or Si-C substructures. In addition to these isosteric exchanges, the development of biologically active compounds bearing unique substructures such as carboranes, hydrophobic boron clusters, and ferrocene is introduced. These novel strategies provide several options for structural development, offering great potential for expanding the chemical space of medicinal chemistry.


Asunto(s)
Bortezomib/síntesis química , Química Farmacéutica/métodos , Diseño de Fármacos/métodos , Descubrimiento de Drogas/métodos , Elementos Químicos , Hidrocarburos/química , Bortezomib/química , Fenómenos Químicos , Química Farmacéutica/tendencias , Halógenos/química , Nitrógeno/química , Oxígeno/química , Fósforo/química , Silicio/química , Azufre/química
6.
Neoplasia ; 24(1): 1-11, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34826777

RESUMEN

The introduction of the proteasome inhibitor bortezomib into treatment regimens for myeloma has led to substantial improvement in patient survival. However, whilst bortezomib elicits initial responses in many myeloma patients, this haematological malignancy remains incurable due to the development of acquired bortezomib resistance. With other patients presenting with disease that is intrinsically bortezomib resistant, it is clear that new therapeutic approaches are desperately required to target bortezomib-resistant myeloma. We have previously shown that targeting sphingolipid metabolism with the sphingosine kinase 2 (SK2) inhibitor K145 in combination with bortezomib induces synergistic death of bortezomib-naïve myeloma. In the current study, we have demonstrated that targeting sphingolipid metabolism with K145 synergises with bortezomib and effectively resensitises bortezomib-resistant myeloma to this proteasome inhibitor. Notably, these effects were dependent on enhanced activation of the unfolded protein response, and were observed in numerous separate myeloma models that appear to have different mechanisms of bortezomib resistance, including a new bortezomib-resistant myeloma model we describe which possesses a clinically relevant proteasome mutation. Furthermore, K145 also displayed synergy with the next-generation proteasome inhibitor carfilzomib in bortezomib-resistant and carfilzomib-resistant myeloma cells. Together, these findings indicate that targeting sphingolipid metabolism via SK2 inhibition may be effective in combination with a broad spectrum of proteasome inhibitors in the proteasome inhibitor resistant setting, and is an approach worth clinical exploration.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Inhibidores de Proteasoma/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Bortezomib/química , Bortezomib/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/uso terapéutico , Técnicas de Inactivación de Genes , Humanos , Ratones , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Inhibidores de Proteasoma/química , Inhibidores de Proteasoma/uso terapéutico , Relación Estructura-Actividad , Respuesta de Proteína Desplegada/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Int J Mol Sci ; 22(23)2021 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-34884766

RESUMEN

Peptides inherently feature the favorable properties of being easily synthesized, water-soluble, biocompatible, and typically non-toxic. Thus, boronic acid has been widely integrated with peptides with the goal of discovering peptide ligands with novel biological activities, and this effort has led to broad applications. Taking the integration between boronic acid and peptide as a starting point, we provide an overview of the latest research advances and highlight the versatile and robust functionalities of boronic acid. In this review, we summarize the diverse applications of peptide boronic acids in medicinal chemistry and chemical biology, including the identification of covalent reversible enzyme inhibitors, recognition, and detection of glycans on proteins or cancer cell surface, delivery of siRNAs, development of pH responsive devices, and recognition of RNA or bacterial surfaces. Additionally, we discuss boronic acid-mediated peptide cyclization and peptide modifications, as well as the facile chemical synthesis of peptide boronic acids, which paved the way for developing a growing number of peptide boronic acids.


Asunto(s)
Ácidos Borónicos/química , Ácidos Borónicos/farmacología , Péptidos/química , Péptidos/farmacología , Antineoplásicos/química , Antineoplásicos/farmacología , Compuestos de Boro/química , Compuestos de Boro/farmacología , Ácidos Borónicos/síntesis química , Bortezomib/química , Bortezomib/farmacología , Diseño de Fármacos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Glicina/análogos & derivados , Glicina/química , Glicina/farmacología , Humanos , Péptidos/síntesis química , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacología
8.
Artículo en Inglés | MEDLINE | ID: mdl-34438277

RESUMEN

Bortezomib, a proteinase inhibitor currently used to treat multiple myeloma and mantle cell lymphoma, has a high incidence of adverse reactions and large inter-individual differences in plasma concentrations. A simple, validated LC-MS/MS method for the quantitative analysis of bortezomib in dried blood spot (DBS) samples was developed to provide support for determining the effective concentration range of bortezomib for clinical use. Fifty (i50) µL of spiked blood were added onto Whatman protein saver cards to prepare the DBS samples. Circular cards of 6 mm diameter were punched, extracted by methanol containing the internal standard (apatinib), and injected into the LC-MS/MS system. The method validation included selectivity, linearity, accuracy and precision, stability, matrix effect, recovery and hematocrit. The calibration curve showed correlation coefficient values higher than 0.999 in the range of 0.2 - 20.0 ng/mL for bortezomib. The acceptance criteria of accuracy (relative error < 12.5%) and precision (coefficient of variation < 10.7%) were met in all cases. The matrix effect was<13.2%, and the recovery was between 87.3 and 100.2%. DBS samples were shown to be stable when stored in cold conditions or at room temperature. Different hematocrit values did not significantly affect the accuracy of the measured concentrations. And there are no significant differences between bortezomib concentrations in DBS samples and plasma samples. This new method was successfully used for clinical concentration determinations of bortezomib and can be applied in future therapeutic drug monitoring and pharmacokinetic studies of bortezomib especially in pediatric patients.


Asunto(s)
Bortezomib/sangre , Cromatografía Liquida/métodos , Pruebas con Sangre Seca/métodos , Espectrometría de Masas en Tándem/métodos , Bortezomib/química , Bortezomib/farmacocinética , Humanos , Límite de Detección , Modelos Lineales , Reproducibilidad de los Resultados
9.
ACS Appl Mater Interfaces ; 13(31): 36926-36937, 2021 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-34319074

RESUMEN

Synergistic therapy holds promising potential in cancer treatment. Here, the inclusion of catechol moieties, a disulfide cross-linked structure, and pendent carboxyl into the network of polymeric nanogels with glutathione (GSH)-responsive dissociation and pH-sensitive release is first disclosed for the codelivery of doxorubicin (DOX) and bortezomib (BTZ) in synergistic cancer therapy. The pendent carboxyl groups and catechol moieties are exploited to absorb DOX through electrostatic interaction and conjugate BTZ through boronate ester, respectively. Both electrostatic interactions and boronate ester are stable at neutral or alkaline pH, while they are instable in an acidic environment to further recover the activities of BTZ and DOX. The polymeric nanogels possess a superior stability to prevent the premature leakage of drugs in a physiological environment, while their structure is destroyed in response to a typical endogenous stimulus (GSH) to unload drugs. The dissociation of the drug-loaded nanogels accelerates the intracellular release of DOX and BTZ and further enhances the therapeutic efficacy. In vitro and in vivo investigations revealed that the dual-drug loaded polymeric nanogels exhibited a strong ability to suppress tumor growth. This study thus proposes a new perspective on the production of multifunctional polymeric nanogels through the introduction of different functional monomers.


Asunto(s)
Antineoplásicos/uso terapéutico , Bortezomib/uso terapéutico , Doxorrubicina/uso terapéutico , Portadores de Fármacos/química , Nanogeles/química , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos/química , Bortezomib/química , Catecoles/química , Cistamina/análogos & derivados , Cistamina/metabolismo , Doxorrubicina/química , Combinación de Medicamentos , Sinergismo Farmacológico , Femenino , Glutatión/metabolismo , Humanos , Células MCF-7 , Ratones Endogámicos NOD , Ratones SCID , Neoplasias/metabolismo , Neoplasias/patología , Polímeros/química , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Int J Biol Macromol ; 183: 369-378, 2021 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-33932413

RESUMEN

Hydrophobic drugs loaded nanogels were always associated with low encapsulation efficiency and immature burst release. In this work, dopamine grafted hyaluronate nanogels were designed for bortezomib (BTZ), a hydrophobic anticancer drug and a proteasome inhibitor. It was found that there was a more efficient loading and pH-controlled release of BTZ due to the presence of dopamine groups on the skeleton of the nanogels. The drug loading content (DLC) were up to 8.58% as the nanogels modified with 29% dopamine, compared to the DLC of less than 1% for nanogels without dopamine modification. It was the pH-sensitive nature of the borated bonds between BTZ and catechol groups that endowed the pH-responsive release behavior of BTZ in vitro. In vitro study proved good biocompatibility and efficient cell uptake of the nanogels. In vivo anti-tumor experiments demonstrated that bortezomib loading into the nanogel significantly enhanced the therapeutic effect of the drug. After 14-day treatment, the average tumor volume of BTZ loaded nanogel group was reduced by 200% more than that of free BTZ group. Combined with CD44 receptor targeting ability of hyaluronate and the merits of nanogel, the catechol modified hyaluronate nanogel exhibited as an efficient chemotherapeutic formulation of BTZ for cancer treatment.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/uso terapéutico , Bortezomib/química , Bortezomib/uso terapéutico , Dopamina/química , Nanogeles/química , Polietilenglicoles/química , Polietileneimina/química , Animales , Portadores de Fármacos/química , Humanos , Concentración de Iones de Hidrógeno
11.
J Med Chem ; 64(8): 4857-4869, 2021 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-33821636

RESUMEN

LONP1 is an AAA+ protease that maintains mitochondrial homeostasis by removing damaged or misfolded proteins. Elevated activity and expression of LONP1 promotes cancer cell proliferation and resistance to apoptosis-inducing reagents. Despite the importance of LONP1 in human biology and disease, very few LONP1 inhibitors have been described in the literature. Herein, we report the development of selective boronic acid-based LONP1 inhibitors using structure-based drug design as well as the first structures of human LONP1 bound to various inhibitors. Our efforts led to several nanomolar LONP1 inhibitors with little to no activity against the 20S proteasome that serve as tool compounds to investigate LONP1 biology.


Asunto(s)
Proteasas ATP-Dependientes/antagonistas & inhibidores , Diseño de Fármacos , Proteínas Mitocondriales/antagonistas & inhibidores , Inhibidores de Proteasas/química , Proteasas ATP-Dependientes/metabolismo , Sitios de Unión , Ácidos Borónicos/química , Ácidos Borónicos/metabolismo , Ácidos Borónicos/farmacología , Bortezomib/química , Bortezomib/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Humanos , Proteínas Mitocondriales/metabolismo , Simulación del Acoplamiento Molecular , Inhibidores de Proteasas/metabolismo , Inhibidores de Proteasas/farmacología , Complejo de la Endopetidasa Proteasomal/química , Complejo de la Endopetidasa Proteasomal/metabolismo , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/metabolismo , Relación Estructura-Actividad
12.
Anticancer Agents Med Chem ; 21(1): 20-32, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32781973

RESUMEN

BACKGROUND: The ubiquitin-proteasome pathway is involved in almost all cellular processes (cell cycle, gene transcription and translation, cell survival and apoptosis, cell metabolism and protein quality control) mainly through the specific degradation of the majority of intracellular proteins (>80%) or partial processing of transcription factors (e.g., NF-κB). A growing amount of evidence now indicates that epigenetic changes are also regulated by the ubiquitin-proteasome pathway. Recent studies indicate that epigenetic regulations are equally crucial for almost all biological processes as well as for pathological conditions such as tumorigenesis, as compared to non-epigenetic control mechanisms (i.e., genetic alterations or classical signal transduction pathways). OBJECTIVE: Here, we reviewed the recent work highlighting the interaction of the ubiquitin-proteasome pathway components (e.g., ubiquitin, E1, E2 and E3 enzymes and 26S proteasome) with epigenetic regulators (histone deacetylases, histone acetyltransferases and DNA methyltransferases). RESULTS: Alterations in the regulation of the ubiquitin-proteasome pathway have been discovered in many pathological conditions. For example, a 2- to 32-fold increase in proteasomal activity and/or subunits has been noted in primary breast cancer cells. Although proteasome inhibitors have been successfully applied in the treatment of hematological malignancies (e.g., multiple myeloma), the clinical efficacy of the proteasomal inhibition is limited in solid cancers. Interestingly, recent studies show that the ubiquitin-proteasome and epigenetic pathways intersect in a number of ways through the regulation of epigenetic marks (i.e., acetylation, methylation and ubiquitylation). CONCLUSION: It is therefore believed that novel treatment strategies involving new generation ubiquitinproteasome pathway inhibitors combined with DNA methyltransferase, histone deacetylase or histone acetyltransferase inhibitors may produce more effective results with fewer adverse effects in cancer treatment as compared to standard chemotherapeutics in hematological as well as solid cancers.


Asunto(s)
Epigénesis Genética/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/química , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Ubiquitina/metabolismo , Acetilación , Compuestos de Boro/farmacología , Bortezomib/química , Bortezomib/farmacología , Metilasas de Modificación del ADN/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Glicina/análogos & derivados , Glicina/farmacología , Histona Acetiltransferasas/antagonistas & inhibidores , Histona Desacetilasas/metabolismo , Humanos , Ácidos Hidroxámicos/farmacología , Metilación , FN-kappa B/metabolismo , Inhibidores de Proteasoma/metabolismo , Inhibidores de Proteasoma/farmacología , Transducción de Señal , Compuestos de Terfenilo/farmacología , Ubiquitinación , Ácido Valproico/farmacología
13.
Cell Mol Life Sci ; 78(3): 1113-1129, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32607595

RESUMEN

Protein homeostasis is essential for life in eukaryotes. Organisms respond to proteotoxic stress by activating heat shock transcription factors (HSFs), which play important roles in cytoprotection, longevity and development. Of six human HSFs, HSF1 acts as a proteostasis guardian regulating stress-induced transcriptional responses, whereas HSF2 has a critical role in development, in particular of brain and reproductive organs. Unlike HSF1, that is a stable protein constitutively expressed, HSF2 is a labile protein and its expression varies in different tissues; however, the mechanisms regulating HSF2 expression remain poorly understood. Herein we demonstrate that the proteasome inhibitor anticancer drug bortezomib (Velcade), at clinically relevant concentrations, triggers de novo HSF2 mRNA transcription in different types of cancers via HSF1 activation. Similar results were obtained with next-generation proteasome inhibitors ixazomib and carfilzomib, indicating that induction of HSF2 expression is a general response to proteasome dysfunction. HSF2-promoter analysis, electrophoretic mobility shift assays, and chromatin immunoprecipitation studies unexpectedly revealed that HSF1 is recruited to a heat shock element located at 1.397 bp upstream from the transcription start site in the HSF2-promoter. More importantly, we found that HSF1 is critical for HSF2 gene transcription during proteasome dysfunction, representing an interesting example of transcription factor involved in controlling the expression of members of the same family. Moreover, bortezomib-induced HSF2 was found to localize in the nucleus, interact with HSF1, and participate in bortezomib-mediated control of cancer cell migration. The results shed light on HSF2-expression regulation, revealing a novel level of HSF1/HSF2 interplay that may lead to advances in pharmacological modulation of these fundamental transcription factors.


Asunto(s)
Factores de Transcripción del Choque Térmico/metabolismo , Proteínas de Choque Térmico/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Factores de Transcripción/metabolismo , Compuestos de Boro/química , Compuestos de Boro/metabolismo , Bortezomib/química , Bortezomib/metabolismo , Bortezomib/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Glicina/análogos & derivados , Glicina/química , Glicina/metabolismo , Factores de Transcripción del Choque Térmico/antagonistas & inhibidores , Factores de Transcripción del Choque Térmico/genética , Proteínas de Choque Térmico/antagonistas & inhibidores , Proteínas de Choque Térmico/genética , Humanos , Regiones Promotoras Genéticas , Complejo de la Endopetidasa Proteasomal/química , Inhibidores de Proteasoma/química , Inhibidores de Proteasoma/metabolismo , Inhibidores de Proteasoma/farmacología , Interferencia de ARN , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Sitio de Iniciación de la Transcripción , Transcripción Genética
14.
Clin Cancer Res ; 26(22): 5952-5961, 2020 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-32913136

RESUMEN

PURPOSE: Proteasome inhibitors are widely used in treating multiple myeloma, but can cause serious side effects and response varies among patients. It is, therefore, important to gain more insight into which patients will benefit from proteasome inhibitors. EXPERIMENTAL DESIGN: We introduce simulated treatment learned signatures (STLsig), a machine learning method to identify predictive gene expression signatures. STLsig uses genetically similar patients who have received an alternative treatment to model which patients will benefit more from proteasome inhibitors than from an alternative treatment. STLsig constructs gene networks by linking genes that are synergistic in their ability to predict benefit. RESULTS: In a dataset of 910 patients with multiple myeloma, STLsig identified two gene networks that together can predict benefit to the proteasome inhibitor, bortezomib. In class "benefit," we found an HR of 0.47 (P = 0.04) in favor of bortezomib, while in class "no benefit," the HR was 0.91 (P = 0.68). Importantly, we observed a similar performance (HR class benefit, 0.46; P = 0.04) in an independent patient cohort. Moreover, this signature also predicts benefit for the proteasome inhibitor, carfilzomib, indicating it is not specific to bortezomib. No equivalent signature can be found when the genes in the signature are excluded from the analysis, indicating that they are essential. Multiple genes in the signature are linked to working mechanisms of proteasome inhibitors or multiple myeloma disease progression. CONCLUSIONS: STLsig can identify gene signatures that could aid in treatment decisions for patients with multiple myeloma and provide insight into the biological mechanism behind treatment benefit.


Asunto(s)
Redes Reguladoras de Genes/efectos de los fármacos , Terapia Molecular Dirigida , Mieloma Múltiple/tratamiento farmacológico , Inhibidores de Proteasoma/química , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Bortezomib/química , Bortezomib/uso terapéutico , Línea Celular Tumoral , Simulación por Computador , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Humanos , Aprendizaje Automático , Mieloma Múltiple/patología , Oligopéptidos/química , Oligopéptidos/uso terapéutico , Complejo de la Endopetidasa Proteasomal/química , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Inhibidores de Proteasoma/uso terapéutico
15.
Science ; 369(6504)2020 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-32764038

RESUMEN

Sulfolobus acidocaldarius is the closest experimentally tractable archaeal relative of eukaryotes and, despite lacking obvious cyclin-dependent kinase and cyclin homologs, has an ordered eukaryote-like cell cycle with distinct phases of DNA replication and division. Here, in exploring the mechanism of cell division in S. acidocaldarius, we identify a role for the archaeal proteasome in regulating the transition from the end of one cell cycle to the beginning of the next. Further, we identify the archaeal ESCRT-III homolog, CdvB, as a key target of the proteasome and show that its degradation triggers division by allowing constriction of the CdvB1:CdvB2 ESCRT-III division ring. These findings offer a minimal mechanism for ESCRT-III-mediated membrane remodeling and point to a conserved role for the proteasome in eukaryotic and archaeal cell cycle control.


Asunto(s)
Proteínas Arqueales/fisiología , División Celular , Complejos de Clasificación Endosomal Requeridos para el Transporte/fisiología , Complejo de la Endopetidasa Proteasomal/fisiología , Sulfolobus acidocaldarius/citología , Proteínas Arqueales/química , Bortezomib/química , Bortezomib/farmacología , Complejos de Clasificación Endosomal Requeridos para el Transporte/química , Modelos Moleculares , Complejo de la Endopetidasa Proteasomal/química , Inhibidores de Proteasoma/química , Inhibidores de Proteasoma/farmacología , Proteolisis , Sulfolobus acidocaldarius/efectos de los fármacos , Sulfolobus acidocaldarius/enzimología
16.
Angew Chem Int Ed Engl ; 59(38): 16445-16450, 2020 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-32521103

RESUMEN

Herein, we show that an enzymatic reaction can generate peptide assemblies that sequestrate proteins to selectively kill cancer cells. A phosphopeptide bearing the antagonistic motif (AVPI) to the inhibitors of apoptotic proteins (IAPs) enters cancer cells and normal cells by caveolin-dependent endocytosis and macropinocytosis, respectively. The AVPI-bearing peptide assemblies sequestrates IAPs and releases bortezomib (BTZ), a proteasome inhibitor, in the cytosol of cancer cells, but rescues the normal cells (namely, HS-5 cells) by trafficking the BTZ into lysosomes. Alkaline phosphatase (ALP) acts as a context-dependent signal for trafficking the peptide/BTZ assemblies and selectively induces the death of the cancer cells. The assemblies of AVPI exhibit enhanced proteolytic resistance. This work, which utilizes the difference in endocytic uptake of enzymatically formed peptide assemblies to selectively kill cancer cells, promises a new way to develop selective cancer therapeutics.


Asunto(s)
Fosfatasa Alcalina/metabolismo , Antineoplásicos/farmacología , Bortezomib/farmacología , Péptidos/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología , Antineoplásicos/química , Antineoplásicos/metabolismo , Bortezomib/química , Bortezomib/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Células HeLa , Humanos , Estructura Molecular , Péptidos/química , Péptidos/metabolismo , Inhibidores de Proteasoma/química , Inhibidores de Proteasoma/metabolismo , Células Tumorales Cultivadas
17.
Eur J Med Chem ; 199: 112367, 2020 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-32474350

RESUMEN

Polymer-drug conjugates synthesized by binding therapeutic agents to functional polymers have long been a mainstay of prodrugs, while the slow drug release, insufficient efficacy of a single drug, and low selectivity hamper the clinical translation. By rational prodrug design, a targeted dual-acidity-labile polysaccharide-di-drugs conjugate was synthesized by one-pot simultaneous Schiff base and boronic esterification reactions between oxidized dextran (Dex-CHO) and cyclo-(Arg-Gly-Asp-D-Phe-Lys) (c(RGDfK)), doxorubicin (DOX), and bortezomib (BTZ). The polysaccharide-di-drugs conjugate (Dex-g-(DOX+BTZ)/cRGD) self-assembled into micelle with a diameter at around 80 nm and released the drugs simultaneously triggered by the acidic conditions. Dex-g-(DOX+BTZ)/cRGD specifically recognized and entered the cancer cells through the RGD-αvß3 integrin interplay, selectively released DOX and BTZ in the acidic intracellular microenvironment, and efficiently inhibited the cell proliferation in vitro. More importantly, Dex-DOX/BTZ/cRGD showed higher intratumoral accumulation and better antitumor efficacy in vivo compared with free drugs and non-targeted control prodrug Dex-g-(DOX+BTZ). The findings indicated that this study provided a facile strategy to develop smart polymer-multi-drugs conjugates for targeted cancer chemotherapy.


Asunto(s)
Antineoplásicos/farmacología , Bortezomib/farmacología , Dextranos/farmacología , Doxorrubicina/farmacología , Melanoma/tratamiento farmacológico , Polisacáridos/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Bortezomib/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Dextranos/química , Relación Dosis-Respuesta a Droga , Doxorrubicina/química , Liberación de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Concentración de Iones de Hidrógeno , Masculino , Melanoma/metabolismo , Melanoma/patología , Ratones , Ratones Endogámicos BALB C , Estructura Molecular , Polisacáridos/química , Relación Estructura-Actividad
18.
J Nanobiotechnology ; 18(1): 57, 2020 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-32245495

RESUMEN

BACKGROUNDS: Intolerable toxicity and unsatisfactory therapeutic effects are still big problems retarding the use of chemotherapy against cancer. Nano-drug delivery system promised a lot in increasing the patients' compliance and therapeutic efficacy. As a unique nano-carrier, supermolecular aggregation nanovehicle has attracted increasing interests due to the following advantages: announcing drug loading efficacy, pronouncing in vivo performance and simplified production process. METHODS: In this study, the supermolecular aggregation nanovehicle of bortezomib (BTZ) was prepared to treat breast cancer. RESULTS: Although many supermolecular nanovehicles are inclined to disintegrate due to the weak intermolecular interactions among the components, the BTZ supermolecules are satisfying stable. To shed light on the reasons behind this, the forces driving the formation of the nanovehicles were detailed investigated. In other words, the interactions among BTZ and other two components were studied to characterize the nanovehicles and ensure its stability. CONCLUSIONS: Due to the promising tumor targeting ability of the BTZ nanovehicles, the supermolecule displayed promising tumor curing effects and negligible systemic toxicity.


Asunto(s)
Antineoplásicos/farmacología , Bortezomib/química , Bortezomib/farmacología , Sistemas de Liberación de Medicamentos/métodos , Animales , Línea Celular Tumoral , Femenino , Humanos , Ensayo de Materiales , Ratones , Ratones Endogámicos BALB C , Nanopartículas , Propiedades de Superficie
19.
Int J Pharm ; 579: 119173, 2020 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-32097684

RESUMEN

Bortezomib (BTZ) is a proteasome inhibitor as approved by US FDA for the treatment of multiple myeloma. It exhibits significant anti-cancer properties, against solid tumors; but lacks aqueous solubility, chemical stability which hinders its successful formulation development. The present study is an attempt to deliver BTZ using N-(2-hydroxypropyl) methacrylamide (HPMA) based copolymeric conjugates and biotinylated PNPs in an effective manner. Study describes a systematic synthetic pathway to synthesize functional polymeric conjugates such as HPMA-Biotin (HP-BT) HPMA-Polylactic acid (HPLA) and HPMA-PLA-Biotin (HPLA-BT) followed by exhaustive characterization both spectroscopically and microscopically. Our strategy yielded polymeric nanoparticles (PNPs) of narrow size range of 199.7 ± 1.32 nm. Release studies were performed at pH 7.4 and 5.6. PNPs were 2-folds less hemolytic (p < 0.0001) than pure drug. BTZ loaded PNPs of HPLA-BT demonstrated significant anti-cancer activity against MCF-7 cells. IC50 value of these PNPs was 56.06 ± 0.12 nM, which was approximately two folds less than BTZ (p < 0.0001). Cellular uptake study confirmed that higher uptake of formulations might be an outcome of biotin surface tethering characteristics that enhanced selectivity and targeting of formulations efficiently. In vivo pharmacokinetics evidenced increased bioavailability (AUC0 t-∞) of DL-HPLA-BT PNPs (drug loaded) than BTZ with an improved half-life. Overall the developed PNPs led to the improved and effective BTZ delivery.


Asunto(s)
Biotinilación/métodos , Bortezomib/química , Sistemas de Liberación de Medicamentos/métodos , Metacrilatos/química , Nanopartículas/química , Polímeros/química , Animales , Disponibilidad Biológica , Bortezomib/efectos adversos , Bortezomib/farmacocinética , Bortezomib/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Liberación de Fármacos , Humanos , Concentración de Iones de Hidrógeno , Tamaño de la Partícula , Ratas
20.
Mar Drugs ; 18(1)2020 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-31947850

RESUMEN

We performed a high-content screening (HCS) assay aiming to discover bioactive molecules with proteasome inhibitory activity. By structural elucidation, we identified six compounds purified from soft coral Clavularia flava, which potentiates proteasome inhibition. Chemical structure elucidation revealed they are dolabellane- and secosteroid-based compounds including a new dolabellane, clavinflol C (1), three known dolabellanes, stolonidiol (2), stolonidiol-17-acetate (3), and clavinflol B (4) as well as two new secosteroids, 3,11-dihydroxy-24-methyl-9,11-secocholest-5-en-9,23-dione (5) and 3,11-dihydroxy-24-methylene-9,11-secocholest-5-en-9,23-dione (6). All six compounds show less cytotoxicity than those of known proteasome inhibitors, bortezomib and MG132. In summary, the high-content measurements of control inhibitors, bortezomib and MG132, manifest the highest ratio >2 in high-content measurement. Of the isolated compounds, 2 and 5 showed higher activity, followed by 3 and 6, and then 1 and 4 exhibited moderate inhibition.


Asunto(s)
Antozoos/química , Diterpenos/química , Complejo de la Endopetidasa Proteasomal/química , Secoesteroides/química , Ubiquitina/química , Animales , Bortezomib/química , Inhibidores de Proteasoma/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA