Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Pharmacol Res ; 204: 107215, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38744399

RESUMO

The ubiquitinproteasome system (UPS) is the main mechanism responsible for the intracellular degradation of misfolded or damaged proteins. Under inflammatory conditions, the immunoproteasome, an isoform of the proteasome, can be induced, enhancing the antigen-presenting function of the UPS. Furthermore, the immunoproteasome also serves nonimmune functions, such as maintaining protein homeostasis and regulating signalling pathways, and is involved in the pathophysiological processes of various cardiovascular diseases (CVDs). This review aims to provide a comprehensive summary of the current research on the involvement of the immunoproteasome in cardiovascular diseases, with the ultimate goal of identifying novel strategies for the treatment of these conditions.


Assuntos
Doenças Cardiovasculares , Complexo de Endopeptidases do Proteassoma , Humanos , Complexo de Endopeptidases do Proteassoma/metabolismo , Complexo de Endopeptidases do Proteassoma/imunologia , Doenças Cardiovasculares/imunologia , Doenças Cardiovasculares/metabolismo , Animais , Ubiquitina/metabolismo , Ubiquitina/imunologia , Transdução de Sinais
2.
Free Radic Biol Med ; 212: 336-348, 2024 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-38176476

RESUMO

Ferroptosis is involved in the pathogenesis of osteoarthritis (OA) while suppression of chondrocyte ferroptosis has a beneficial effect on OA. However, the molecular mechanism of ferroptosis in OA remains to be elucidated. P21, an indicator of aging, has been reported to inhibit ferroptosis, but the relationship between P21 and ferroptosis in OA remains unclear. Here, we aimed to investigate the expression and function of P21 in OA chondrocytes, and the involvement of P21 in the regulation of ferroptosis in chondrocytes. First, we demonstrated that high P21 expression was observed in the cartilage from OA patients and destabilized medial meniscus (DMM) mice, and in osteoarthritic chondrocytes induced by IL-1ß, FAC and erastin. P21 knockdown exacerbated the reduction of Col2a1 and promoted the upregulation of MMP13 in osteoarthritic chondrocytes. Meanwhile, P21 knockdown exacerbated cartilage degradation in DMM-induced OA mouse models and decreased GPX4 expression in vivo. Furthermore, P21 knockdown sensitized chondrocytes to ferroptosis induced by erastin, which was closely associated with the accumulation of lipid peroxides. In mechanism, we demonstrated that P21 regulated the stability of GPX4 protein, and the regulation was independent of NRF2. Meanwhile, we found that P21 significantly affected the recruitment of GPX4 to linear ubiquitin chain assembly complex (LUBAC) and regulated the level of M1-linked ubiquitination of GPX4. Overall, our results suggest that P21 plays an essential anti-ferroptosis role in OA by regulating the stability of GPX4.


Assuntos
Ferroptose , Osteoartrite , Humanos , Camundongos , Animais , Condrócitos/metabolismo , Ferroptose/genética , Cartilagem/metabolismo , Modelos Animais de Doenças , Regulação para Cima , Osteoartrite/genética , Osteoartrite/metabolismo
3.
Pharmacol Res ; 194: 106830, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37343647

RESUMO

Drug combination therapy is a highly effective approach for enhancing the therapeutic efficacy of anti-cancer drugs and overcoming drug resistance. However, the innumerable possible drug combinations make it impractical to screen all synergistic drug pairs. Moreover, biological insights into synergistic drug pairs are still lacking. To address this challenge, we systematically analyzed drug combination datasets curated from multiple databases to identify drug pairs more likely to show synergy. We classified drug pairs based on their MoA and discovered that 110 MoA pairs were significantly enriched in synergy in at least one type of cancer. To improve the accuracy of predicting synergistic effects of drug pairs, we developed a suite of machine learning models that achieve better predictive performance. Unlike most previous methods that were rarely validated by wet-lab experiments, our models were validated using two-dimensional cell lines and three-dimensional tumor slice culture (3D-TSC) models, implying their practical utility. Our prediction and validation results indicated that the combination of the RTK inhibitors Lapatinib and Pazopanib exhibited a strong therapeutic effect in breast cancer by blocking the downstream PI3K/AKT/mTOR signaling pathway. Furthermore, we incorporated molecular features to identify potential biomarkers for synergistic drug pairs, and almost all potential biomarkers found connections between drug targets and corresponding molecular features using protein-protein interaction network. Overall, this study provides valuable insights to complement and guide rational efforts to develop drug combination treatments.


Assuntos
Antineoplásicos , Neoplasias da Mama , Humanos , Feminino , Fosfatidilinositol 3-Quinases , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Transdução de Sinais , Neoplasias da Mama/tratamento farmacológico , Sistemas de Liberação de Medicamentos
4.
Biochem Pharmacol ; 204: 115232, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36030831

RESUMO

ONC201 is a well-known caseinolytic protease (ClpP)activator with established benefits against multiple tumors, including colorectal cancer (CRC). In this study, we investigated the anticancer effects and associated mechanisms of the ClpP agonist IMP075, derived from ONC201. Acute toxicity and CCK-8 assayswere employed to determine the safety of IMP075. The effectiveness of IMP075 was investigated in HCT116 cells and a mouse xenograft tumor model. Additionally, the properties of IMP075 were evaluated by pharmacokinetic,CYP inhibition, and hERG inhibition assays. Finally, isothermal titration calorimetry (ITC), differential scanning fluorimetry (DSF), cellular thermal shift assay (CETSA), molecular dynamics simulations, point mutations, and shRNA experiments were employed to elucidate the potential mechanism of IMP075. Compared with ONC201, IMP075 exhibited similar toxicity and improved antitumor effects in vitro and in vivo. Interestingly, the affinity and agonistic effects of IMP075 on ClpP were superior to ONC201, which allowed IMP075 to disrupt respiratory chain integrity at lower doses in HCT116 cells, leading to mitochondrial dysfunction. Furthermore, molecular dynamics simulations demonstrate that IMP075 forms two pairs of hydrogen bonds with ClpP, maintaining ClpP in an agonistic state. Importantly, the antiproliferative activity of IMP075 significantly decreased following ClpP knockdown. Our findings substantiate that IMP075 exerts excellent antitumor effects against CRC by activating ClpP-mediated impairment of mitochondrial function. Due to its superior properties, IMP075 appears to be have huge prospects for application.


Assuntos
Neoplasias do Colo , Peptídeo Hidrolases , Animais , Linhagem Celular Tumoral , Neoplasias do Colo/tratamento farmacológico , Endopeptidase Clp/genética , Endopeptidase Clp/metabolismo , Humanos , Camundongos , Peptídeo Hidrolases/uso terapêutico , RNA Interferente Pequeno , Sincalida/uso terapêutico
5.
Pharmacol Res ; 163: 105248, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33065283

RESUMO

The ubiquitin-proteasome system constitutes a major pathway for protein degradation in the cell. Therefore the crosstalk of this pathway with mitochondria is a major topic with direct relevance to many mitochondrial diseases. Proteasome dysfunction triggers not only protein toxicity, but also mitochondrial dysfunction. The involvement of proteasomes in the regulation of protein transport into mitochondria contributes to an increase in mitochondrial function defects. On the other hand, mitochondrial impairment stimulates reactive oxygen species production, which increases protein damage, and protein misfolding and aggregation leading to proteasome overload. Concurrently, mitochondrial dysfunction compromises cellular ATP production leading to reduced protein ubiquitination and proteasome activity. In this review we discuss the complex relationship and interdependence of the ubiquitin-proteasome system and mitochondria. Furthermore, we describe pharmacological inhibition of proteasome activity as a novel strategy to treat a group of mitochondrial diseases.


Assuntos
Mitocôndrias/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo , Animais , Humanos , Doenças Mitocondriais/tratamento farmacológico , Doenças Mitocondriais/metabolismo , Peptídeos/metabolismo
6.
Pharmacol Res ; 161: 105218, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33007418

RESUMO

Endoplasmic reticulum (ER) stress is easily observed in chronic liver disease, which often causes accumulation of unfolded or misfolded proteins in the ER, leading to unfolded protein response (UPR). Regulating protein degradation is an integral part of UPR to relieve ER stress. The major protein degradation system includes the ubiquitin-proteasome system (UPS) and autophagy. All three arms of UPR triggered in response to ER stress can regulate UPS and autophagy. Accumulated misfolded proteins could activate these arms, and then generate various transcription factors to regulate the expression of UPS-related and autophagy-related genes. The protein degradation process regulated by UPR has great significance in many chronic liver diseases, including non-alcoholic fatty liver disease (NAFLD), alcoholic liver disease (ALD), viral hepatitis, liver fibrosis, and hepatocellular carcinoma(HCC). In most instances, the degradation of excessive proteins protects cells with ER stress survival from apoptosis. According to the specific functions of protein degradation in chronic liver disease, choosing to promote or inhibit this process is promising as a potential method for treating chronic liver disease.


Assuntos
Estresse do Retículo Endoplasmático , Hepatopatias/metabolismo , Fígado/metabolismo , Proteostase , Animais , Autofagia , Doença Crônica , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Humanos , Fígado/efeitos dos fármacos , Fígado/patologia , Hepatopatias/tratamento farmacológico , Hepatopatias/patologia , Proteólise , Proteostase/efeitos dos fármacos , Resposta a Proteínas não Dobradas
7.
Pharmacol Res ; 161: 105143, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32814168

RESUMO

Stress granules (SGs) are assemblies of mRNA and proteins that form from mRNAs stalled in translation initiation in response to stress. Chronic stress might even induce formation of cytotoxic pathological SGs. SGs participate in various biological functions including response to apoptosis, inflammation, immune modulation, and signalling pathways; moreover, SGs are involved in pathogenesis of neurodegenerative diseases, viral infection, aging, cancers and many other diseases. Emerging evidence has shown that small molecules can affect SG dynamics, including assembly, disassembly, maintenance and clearance. Thus, targeting SGs is a potential therapeutic strategy for the treatment of human diseases and the promotion of health. The established methods for detecting SGs provided ready tools for large-scale screening of agents that alter the dynamics of SGs. Here, we describe the effects of small molecules on SG assembly, disassembly, and their roles in the disease. Moreover, we provide perspective for the possible application of small molecules targeting SGs in the treatment of human diseases.


Assuntos
Antineoplásicos/farmacologia , Antivirais/farmacologia , Grânulos Citoplasmáticos/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Doenças Neurodegenerativas/tratamento farmacológico , Proteínas/metabolismo , RNA Mensageiro/metabolismo , Estresse Fisiológico , Viroses/tratamento farmacológico , Envelhecimento , Animais , Grânulos Citoplasmáticos/genética , Grânulos Citoplasmáticos/metabolismo , Grânulos Citoplasmáticos/patologia , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , RNA Mensageiro/genética , Transdução de Sinais , Viroses/genética , Viroses/metabolismo , Viroses/patologia
8.
Biomed Pharmacother ; 128: 110330, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32504922

RESUMO

MEF2D-fusion (M-fusion) genes are newly discovered recurrent gene abnormalities that are detected in approximately 5 % of acute lymphoblastic leukemia (ALL) cases. Their introduction to cells has been reported to transform cell lines or increase the colony formation of bone marrow cells, suggesting their survival-supporting ability, which prompted us to examine M-fusion-targeting drugs. To identify compounds that reduce the protein expression level of MEF2D, we developed a high-throughput screening system using 293T cells stably expressing a fusion protein of MEF2D and luciferase, in which the protein expression level of MEF2D was easily measured by a luciferase assay. We screened 3766 compounds with known pharmaceutical activities using this system and selected staurosporine as a potential inducer of the proteolysis of MEF2D. Staurosporine induced the proteolysis of M-fusion proteins in M-fusion (+) ALL cell lines. Proteolysis was inhibited by caspase inhibitors, not proteasome inhibitors, suggesting caspase dependency. Consistent with this result, the growth inhibitory effects of staurosporine were stronger in M-fusion (+) ALL cell lines than in negative cell lines, and caspase inhibitors blocked apoptosis induced by staurosporine. We identified the cleavage site of MEF2D-HNRNPUL1 by caspases and confirmed that its caspase cleavage-resistant mutant was resistant to staurosporine-induced proteolysis. Based on these results, we investigated another Food and Drug Administration-approved caspase activator, venetoclax, and found that it exerted similar effects to staurosporine, namely, the proteolysis of M-fusion proteins and strong growth inhibitory effects in M-fusion (+) ALL cell lines. The present study provides novel insights into drug screening strategies and the clinical indications of venetoclax.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Caspases/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Estaurosporina/farmacologia , Sulfonamidas/farmacologia , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Fusão Gênica , Células HEK293 , Humanos , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Proteólise , Transdução de Sinais
9.
Pharmacol Res ; 155: 104716, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32084560

RESUMO

TNF-related apoptosis-inducing ligand (TRAIL) selectively induces the apoptosis pathway in tumor cells leading to tumor cell death. Because TRAIL induction can kill tumor cells, cancer researchers have developed many agents to target TRAIL and some of these agents have entered clinical trials in oncology. Unfortunately, these trials have failed for many reasons, including drug resistance, off-target toxicities, short half-life, and specifically in gene therapy due to the limited uptake of TRAIL genes by cancer cells. To address these drawbacks, translational researchers have utilized drug delivery platforms. Although, these platforms can improve TRAIL-based therapies, they are unable to sufficiently translate the full potential of TRAIL-targeting to clinically viable products. Herein, we first summarize the complex biology of TRAIL signaling, including TRAILs cross-talk with other signaling pathways and immune cells. Next, we focus on known resistant mechanisms to TRAIL-based therapies. Then, we discuss how nano-formulation has the potential to enhance the therapeutic efficacy of TRAIL protein. Finally, we specify strategies with the potential to overcome the challenges that cannot be addressed via nanotechnology alone, including the alternative methods of TRAIL-expressing circulating cells, tumor-targeting bacteria, viruses, and exosomes.


Assuntos
Neoplasias/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Animais , Humanos , Terapia de Alvo Molecular , Nanomedicina , Neoplasias/tratamento farmacológico , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Proteínas Recombinantes/metabolismo , Transdução de Sinais
10.
Drug Discov Today ; 25(2): 367-379, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31765717

RESUMO

Multiple myeloma (MM), a plasma cell malignancy, is characterised by lesions in multiple bones involving transformed, matured post-follicular B cells. The course of the disease involves an initial development of monoclonal gammopathy of undetermined significance (MGUS), followed by smouldering MM, before the full MM disease emerges. Despite novel therapies, MM remains incurable, managed by combination therapies, including proteasome inhibitors (PIs), immunomodulators (IMiDs) and anti-human CD38 (daratumumab). MM patients have an increased risk of thromboembolic events due to combination treatments with IMiDs, PIs and anti-human CD38 antibody, and steroids. This review will examine the efficacy and pro-thrombotic effects of MM therapies.


Assuntos
Antineoplásicos/uso terapêutico , Mieloma Múltiplo/tratamento farmacológico , Animais , Humanos
11.
Eur J Pharm Sci ; 134: 116-137, 2019 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-30981885

RESUMO

Autophagy is an evolutionarily conserved catabolic mechanism, by which eukaryotic cells recycle or degrades internal constituents through membrane-trafficking pathway. Thus, autophagy provides the cells with a sustainable source of biomolecules and energy for the maintenance of homeostasis under stressful conditions such as tumor microenvironment. Recent findings revealed a close relationship between autophagy and malignant transformation. However, due to the complex dual role of autophagy in tumor survival or cell death, efforts to develop efficient treatment strategies targeting the autophagy/cancer relation have largely been unsuccessful. Here we review the two-faced role of autophagy in cancer as a tumor suppressor or as a pro-oncogenic mechanism. In this sense, we also review the shared regulatory pathways that play a role in autophagy and malignant transformation. Finally, anti-cancer therapeutic agents used as either inhibitors or inducers of autophagy have been discussed.


Assuntos
Autofagia/efeitos dos fármacos , Autofagia/fisiologia , Neoplasias/metabolismo , Animais , Antineoplásicos , Genes Supressores de Tumor , Humanos , Terapia de Alvo Molecular , Neoplasias/terapia , Oncogenes , Microambiente Tumoral
12.
Biochem Pharmacol ; 143: 118-128, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28642038

RESUMO

Dihydropyrimidinase (DHP, EC 3.5.2.2), encoded by the gene DPYS, is the second enzyme in the catabolic pathway of pyrimidine and of fluoropyrimidine drugs such as 5-fluorouracil, which are commonly used in anticancer treatment; DHP catalyzes the hydrolytic ring opening of dihydrouracil and dihydro-5-fluorouracil. DPYS mutations are known to contribute to interindividual variations in the toxicity of fluoropyrimidine drugs, but the functional characterization of DHP allelic variants remains inadequate. In this study, in vitro analysis was performed on 22 allelic variants of DHP by transiently expressing wild-type DHP and 21 DHP variants in 293FT cells and characterizing their enzymatic activities by using dihydrouracil and dihydro-5-fluorouracil as substrates. DHP expression levels and oligomeric forms were determined using immunoblotting and blue native PAGE, respectively, and the stability of the DHP variants was assessed by examining the proteins in variant-transfected cells treated with cycloheximide or bortezomib. Moreover, three kinetic parameters, Km, Vmax, and intrinsic clearance (Vmax/Km), for the hydrolysis of dihydrouracil and dihydro-5-fluorouracil were determined. We found that 5/21 variants showed significantly decreased intrinsic clearance as compared to wild-type DHP, and that 9/21 variants were expressed at low levels and were inactive due to proteasome-mediated degradation. The band patterns observed in the immunoblotting of blue native gels corresponded to DHP activity, and, notably, 18/21 DHP variants exhibited decreased or null enzymatic activity and these variants also showed a drastically reduced ability to form large oligomers. Thus, detection of DPYS genetic polymorphisms might facilitate the prediction severe adverse effects of fluoropyrimidine-based treatments.


Assuntos
Amidoidrolases , Antineoplásicos/metabolismo , Fluoruracila/análogos & derivados , Variação Genética , Uracila/análogos & derivados , Alelos , Amidoidrolases/genética , Amidoidrolases/metabolismo , Linhagem Celular , Relação Dose-Resposta a Droga , Fluoruracila/metabolismo , Humanos , Modelos Moleculares , Estabilidade Proteica , Especificidade por Substrato , Transfecção , Uracila/metabolismo
13.
Biochem Pharmacol ; 137: 29-50, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28476333

RESUMO

The proteasomal 19S regulatory particle (RP) associated deubiquitinases (DUBs) have attracted much attention owing to their potential as a therapeutic target for cancer therapy. Identification of new entities against 19S RP associated DUBs and illustration of the underlying mechanisms is crucial for discovery of novel proteasome blockers. In this study, a series of 4-arylidene curcumin analogues were identified as potent proteasome inhibitor by preferentially blocking deubiquitinase function of proteasomal 19S RP with moderate 20S CP inhibition. The most active compound 33 exhibited a major inhibitory effect on 19S RP-associated ubiquitin-specific proteases 14, along with a minor effect on ubiquitin C-terminal hydrolase 5, which resulted in dysfunction of proteasome, and subsequently accumulated ubiquitinated proteins (such as IκB) in several cancer cells. Remarkably, though both 19S RP and 20S CP inhibition induced significantly endoplasmic reticulum stress and triggered caspase-12/9 pathway activation to promote cancer cell apoptosis, the 19S RP inhibition by 33 avoided slow onset time, Bcl-2 overexpression, and PERK-phosphorylation, which contribute to the deficiencies of clinical drug Bortezomib. These systematic studies provided insights in the development of novel proteasome inhibitors for cancer treatment.


Assuntos
Antineoplásicos/farmacologia , Curcumina/análogos & derivados , Curcumina/farmacologia , Enzimas Desubiquitinantes/antagonistas & inibidores , Complexo de Endopeptidases do Proteassoma , Inibidores de Proteassoma/farmacologia , Células A549 , Animais , Antineoplásicos/química , Células CHO , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Cricetinae , Cricetulus , Enzimas Desubiquitinantes/metabolismo , Relação Dose-Resposta a Droga , Sistemas de Liberação de Medicamentos/métodos , Humanos , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/química
14.
Eur J Pharmacol ; 791: 99-104, 2016 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-27568836

RESUMO

Platelets express key proteins of the proteasome system and contain protein ubiquitination pathways. The functional role of the proteasome system in platelets, however, is still subject of studies. In addition to its role as anticancer drug, the potent and selective proteasome inhibitor bortezomib can be used for experimental proteasome research. Since it is mandatory to know exact dose-effect relationships, we intended to evaluate dose-dependent specific bortezomib effects on basal and on agonist-induced proteasome activitiy, on levels of poly-ubiquitinated proteins and on platelet aggregation. In washed platelets, unstimulated or stimulated with different agonists and pre-incubated with various bortezomib concentrations, the proteasome activity was determined by a fluorometric assay. The levels of poly-ubiquitinated proteins were assessed by an immunoassay kit. Platelet aggregation was measured by light transmission aggregometry in platelet-rich-plasma. Platelet agonists stimulate both, the proteasome activity and the accumulation of poly-ubiquitinated proteins in platelets. Bortezomib inhibits the basal and the agonist induced proteasome activity and increased the content of poly-ubiquitinated proteins in a concentration dependent manner. Bortezomib concentrations in the nM-range causing complete blockade of platelet proteasome activity do not affect agonist induced platelet aggregation, indicating that the level of platelet proteasome activity is not directly linked with the induction of platelet aggregation. Bortezomib in the µM-range may tamper platelet aggregation, possibly due to unspecific and toxic effects.


Assuntos
Plaquetas/efeitos dos fármacos , Bortezomib/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/farmacologia , Plaquetas/metabolismo , Plaquetas/fisiologia , Proteínas Sanguíneas/metabolismo , Relação Dose-Resposta a Droga , Feminino , Humanos , Masculino , Agregação Plaquetária/efeitos dos fármacos , Ubiquitinação/efeitos dos fármacos , Adulto Jovem
15.
Life Sci ; 145: 152-60, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26706287

RESUMO

Recently, investigators have shown that ubiquitin-proteasome-mediated protein degradation is critical in regulating the balance between bone formation and bone resorption. The major signal transduction pathways regulating bone formation are the RANK/NF-κB pathway and the Wnt/ß-catenin pathway. These signal transduction pathways regulate the activity of mature osteoblasts and osteoclasts. In addition, the Wnt/ß-catenin pathway is one of the major signaling pathways in the differentiation of osteoblasts. The ubiquitin ligases that are reported to be of major significance in regulating these pathways are the ubiquitin SCF(B-TrCP) ligase (which regulates activation of NF-κB via degradation of IkBα in osteoclasts, and regulates bone transcription factors via degradation of ß-catenin), the Keap-Cul3-Rbx1 ligase (which regulates degradation of IkB kinase, Nrf2, and the antiapoptotic factor Bcl-2), and Smurf1. Also of significance in regulating osteoclastogenesis is the deubiquitinase, CYLD (cylindramatosis protein), which facilitates the separation of NF-κB from IkBα. The degradation of CYLD is also under the regulation of SCF(B-TrCP). Proteasome inhibitors influence the activity of mature osteoblasts and osteoclasts, but also modulate the differentiation of precursor cells into osteoblasts. Preclinical studies show that melatonin also influences bone metabolism by stimulating bone growth and inhibiting osteoclast activity. These actions of melatonin could be interpreted as being mediated by the ubiquitin ligases SCF(B-TrCP) and Keap-Cul3-Rbx, or as an inhibitory effect on proteasomes. Clinical trials of the use of melatonin in the treatment of bone disease, including multiple myeloma, using both continuous and intermittent modes of administration, are warranted.


Assuntos
Reabsorção Óssea/metabolismo , Melatonina/metabolismo , Osteogênese , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina/metabolismo , Animais , Doenças Ósseas/metabolismo , Doenças Ósseas/patologia , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Ritmo Circadiano , Humanos , NF-kappa B/metabolismo , Hormônio Paratireóideo/metabolismo , Ligante RANK/metabolismo , Proteínas Ligases SKP Culina F-Box/metabolismo , Transdução de Sinais
16.
Pharmacol Res ; 95-96: 138-58, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25829334

RESUMO

Herbal medicine has attracted great attention in the recent years and is increasingly used as alternatives to chemical drugs. Several lines of evidence support the positive impact of medicinal plants in the prevention and cure of a wide range of diseases. Thymoquinone (TQ) is the most abundant constituent of the volatile oil of Nigella sativa seeds and most properties of N sativa are mainly attributed to TQ. A number of pharmacological actions of TQ have been investigated including anti-oxidant, anti-inflammatory, immunomodulatory, anti-histaminic, anti-microbial and anti-tumor effects. It has also gastroprotective, hepatoprotective, nephroprotective and neuroprotective activities. In addition, positive effects of TQ in cardiovascular disorders, diabetes, reproductive disorders and respiratory ailments, as well as in the treatment of bone complications as well as fibrosis have been shown. In addition, a large body of data shows that TQ has very low adverse effects and no serious toxicity. More recently, a great deal of attention has been given to this dietary phytochemical with an increasing interest to investigate it in pre-clinical and clinical researches for assessing its health benefits. Here we report on and analyze numerous properties of the active ingredient of N. sativa seeds, TQ, in the context of its therapeutic potentials for a wide range of illnesses. We also summarize the drug's possible mechanisms of action. The evidence reported sugests that TQ should be developed as a novel drug in clinical trials.


Assuntos
Benzoquinonas/uso terapêutico , Nigella sativa/química , Óleos de Plantas/química , Sementes/química , Animais , Benzoquinonas/efeitos adversos , Benzoquinonas/isolamento & purificação , Benzoquinonas/farmacologia , Humanos
17.
Life Sci ; 115(1-2): 8-14, 2014 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-25219883

RESUMO

Proteasome inhibitors and melatonin are both intimately involved in the regulation of major signal transduction proteins including p53, cyclin p27, transcription factor NF-κB, apoptotic factors Bax and Bim, caspase 3, caspase 9, anti-apoptotic factor Bcl-2, TRAIL, NRF2 and transcription factor beta-catenin. The fact that these factors are shared targets of the proteasome inhibitor bortezomib and melatonin suggests the working hypothesis that melatonin is a proteasome inhibitor. Supporting this hypothesis is the fact that melatonin shares with bortezomib a selective pro-apoptotic action in cancer cells. Furthermore, both bortezomib and melatonin increase the sensitivity of human glioma cells to TRAIL-induced apoptosis. Direct evidence for melatonin inhibition of the proteasome was recently found in human renal cancer cells. We raise the issue whether melatonin should be investigated in combination with proteasome inhibitors to reduce toxicity, to reduce drug resistance, and to enhance efficacy. This may be particularly valid for hematological malignancies in which proteasome inhibitors have been shown to be useful. Further studies are necessary to determine whether the actions of melatonin on cellular signaling pathways are due to a direct inhibitory effect on the catalytic core of the proteasome, due to an inhibitory action on the regulatory particle of the proteasome, or due to an indirect effect of melatonin on phosphorylation of signal transducing factors.


Assuntos
Melatonina/metabolismo , Melatonina/farmacologia , Neoplasias/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/metabolismo , Inibidores de Proteassoma/farmacologia , Animais , Apoptose/efeitos dos fármacos , Ácidos Borônicos/farmacologia , Bortezomib , Caspases/metabolismo , Humanos , NF-kappa B/metabolismo , Neoplasias/tratamento farmacológico , Pirazinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA