Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Aging (Albany NY) ; 15(5): 1279-1305, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36863315

RESUMO

The direct cause of periodontitis is periodontopathic bacteria, while various environmental factors affect the severity of periodontitis. Previous epidemiological studies have shown positive correlations between aging and periodontitis. However, whether and how aging is linked to periodontal health and disease in biological processes is poorly understood. Aging induces pathological alterations in organs, which promotes systemic senescence associated with age-related disease. Recently, it has become evident that senescence at the cellular level, cellular senescence, is a cause of chronic diseases through production of various secretory factors including proinflammatory cytokines, chemokines, and matrix metalloproteinases (MMPs), which is referred to the senescence-associated secretory phenotype (SASP). In this study, we examined the pathological roles of cellular senescence in periodontitis. We found localization of senescent cells in periodontal tissue, particularly the periodontal ligament (PDL), in aged mice. Senescent human PDL (HPDL) cells showed irreversible cell cycle arrest and SASP-like phenotypes in vitro. Additionally, we observed age-dependent upregulation of microRNA (miR)-34a in HPDL cells. These results suggest that chronic periodontitis is mediated by senescent PDL cells that exacerbate inflammation and destruction of periodontal tissues through production of SASP proteins. Thus, miR-34a and senescent PDL cells might be promising therapeutic targets for periodontitis in elderly people.


Assuntos
MicroRNAs , Ligamento Periodontal , Humanos , Animais , Camundongos , Idoso , Ligamento Periodontal/metabolismo , Envelhecimento/fisiologia , Senescência Celular/fisiologia , Inflamação/metabolismo
2.
J Periodontal Res ; 58(1): 175-183, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36494917

RESUMO

BACKGROUND AND OBJECTIVE: Aggressive periodontitis (AgP) is characterized by general health and rapid destruction of periodontal tissue. The familial aggregation of this disease highlights the involvement of genetic factors in its pathogeny. We conducted a genome-wide association study (GWAS) to identify AgP-related genes in a Japanese population, and the lipid metabolism-related gene, lipase-a, lysosomal acid type (LIPA), was suggested as an AgP candidate gene. However, there is no report about the expression and function(s) of LIPA in periodontal tissue. Hence, we studied the involvement of how LIPA and its single-nucleotide polymorphism (SNP) rs143793106 in AgP by functional analyses of LIPA and its SNP in human periodontal ligament (HPDL) cells. MATERIALS AND METHODS: GWAS was performed using the genome database of Japanese AgP patients, and the GWAS result was confirmed using Sanger sequencing. We examined the mRNA expression level of LIPA and the protein expression level of the encoded protein lysosomal acid lipase (LAL) in periodontium-composing cells using conventional and real-time polymerase chain reaction (PCR) and western blotting, respectively. Lentiviral vectors expressing LIPA wild-type (LIPA WT) and LIPA SNP rs143793106 (LIPA mut) were transfected into HPDL cells. Western blotting was performed to confirm the transfection. LAL activity of transfected HPDL cells was determined using the lysosomal acid lipase activity assay. Transfected HPDL cells were cultured in mineralization medium. During the cytodifferentiation of transfected HPDL cells, mRNA expression of calcification-related genes, alkaline phosphatase (ALPase) activity and calcified nodule formation were assessed using real-time PCR, ALPase assay, and alizarin red staining, respectively. RESULTS: The GWAS study identified 11 AgP-related candidate genes, including LIPA SNP rs143793106. The minor allele frequency of LIPA SNP rs143793106 in AgP patients was higher than that in healthy subjects. LIPA mRNA and LAL protein were expressed in HPDL cells; furthermore, they upregulated the cytodifferentiation of HPDL cells. LAL activity was lower in LIPA SNP-transfected HPDL cells during cytodifferentiation than that in LIPA WT-transfected HPDL cells. In addition, ALPase activity, calcified nodule formation, and calcification-related gene expression levels were lower during cytodifferentiation in LIPA SNP-transfected HPDL cells than those in LIPA WT-transfected HPDL cells. CONCLUSION: LIPA, identified as an AgP-related gene in a Japanese population, is expressed in HPDL cells and is involved in regulating cytodifferentiation of HPDL cells. LIPA SNP rs143793106 suppressed cytodifferentiation of HPDL cells by decreasing LAL activity, thereby contributing to the development of AgP.


Assuntos
Periodontite Agressiva , Humanos , Periodontite Agressiva/genética , Periodontite Agressiva/metabolismo , Ligamento Periodontal , Lipase/genética , Lipase/metabolismo , Polimorfismo de Nucleotídeo Único/genética , Estudo de Associação Genômica Ampla , Esterol Esterase/genética , Esterol Esterase/metabolismo , Diferenciação Celular/genética , RNA Mensageiro/metabolismo , Células Cultivadas
3.
Sci Rep ; 11(1): 1291, 2021 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-33446772

RESUMO

Autophagy is a lysosomal protein degradation system in which the cell self-digests its intracellular protein components and organelles. Defects in autophagy contribute to the pathogenesis of age-related chronic diseases, such as myocardial infarction and rheumatoid arthritis, through defects in the extracellular matrix (ECM). However, little is known about autophagy in periodontal diseases characterised by the breakdown of periodontal tissue. Tooth-supportive periodontal ligament (PDL) tissue contains PDL cells that produce various ECM proteins such as collagen to maintain homeostasis in periodontal tissue. In this study, we aimed to clarify the physiological role of autophagy in periodontal tissue. We found that autophagy regulated type I collagen synthesis by elimination of misfolded proteins in human PDL (HPDL) cells. Inhibition of autophagy by E-64d and pepstatin A (PSA) or siATG5 treatment suppressed collagen production in HPDL cells at mRNA and protein levels. Immunoelectron microscopy revealed collagen fragments in autolysosomes. Accumulation of misfolded collagen in HPDL cells was confirmed by sodium dodecyl sulfate-polyacrylamide gel electrophoresis. E-64d and PSA treatment suppressed and rapamycin treatment accelerated the hard tissue-forming ability of HPDL cells. Our findings suggest that autophagy is a crucial regulatory process that facilitates type I collagen synthesis and partly regulates osteoblastic differentiation of PDL cells.


Assuntos
Autofagia , Colágeno Tipo I/metabolismo , Ligamento Periodontal/citologia , Linhagem Celular , Cadeia alfa 1 do Colágeno Tipo I , Humanos , Ligamento Periodontal/metabolismo , Biossíntese de Proteínas
5.
J Cell Physiol ; 234(5): 7149-7160, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30370560

RESUMO

Fibroblast growth factor-2 (FGF-2) stimulates periodontal regeneration by a broad spectrum of effects on periodontal ligament (PDL) cells, such as proliferation, migration, and production of extracellular matrix. A critical factor in the success of periodontal regeneration is the rapid resolution of inflammatory responses in the tissue. We explored an anti-inflammatory effect of FGF-2 during periodontal regeneration and healing. We found that FGF-2 on mouse periodontal ligament cells (MPDL22) markedly downregulated CD40 expression, a key player of inflammation. In addition, FGF-2 inhibited CD40 signaling by the non-canonical nuclear factor-kappa B2 (NFκB2) pathway, resulting in decreased production of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α), which have the potential to recruit immune cells to inflamed sites. Furthermore, in vivo treatment of FGF-2 enhanced healing of skin wounds by counteracting the CD40-mediated inflammation. These results reveal that FGF-2 has an important function as a negative regulator of inflammation during periodontal regeneration and healing.


Assuntos
Anti-Inflamatórios/farmacologia , Antígenos CD40/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Ligamento Periodontal/efeitos dos fármacos , Periodontite/prevenção & controle , Animais , Antígenos CD40/genética , Linhagem Celular , Modelos Animais de Doenças , Interleucina-6/metabolismo , Masculino , Camundongos Endogâmicos BALB C , Subunidade p52 de NF-kappa B/metabolismo , Ligamento Periodontal/metabolismo , Ligamento Periodontal/patologia , Periodontite/genética , Periodontite/metabolismo , Periodontite/patologia , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo , Cicatrização/efeitos dos fármacos , Ferimentos Penetrantes/tratamento farmacológico , Ferimentos Penetrantes/metabolismo , Ferimentos Penetrantes/patologia
6.
J Periodontal Res ; 54(3): 199-206, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30303256

RESUMO

To identify the genetic risk factors for aggressive periodontitis (AgP), it is important to understand the progression and pathogenesis of AgP. The purpose of this review was to summarize the genetic risk factors for AgP identified through a case-control genomewide association study (GWAS) and replication study. The initial studies to identify novel AgP risk factors were potentially biased because they relied on previous studies. To overcome this kind of issue, an unbiased GWAS strategy was introduced to identify genetic risk factors for various diseases. Currently, three genes glycosyltransferase 6 domain containing 1 (GLT6D1), defensin α1 and α3 (DEFA1A3), and sialic acid-binding Ig-like lectin 5 (SIGLEC5) that reach the threshold for genomewide significance have been identified as genetic risk factors for AgP through a case-control GWAS.


Assuntos
Periodontite Agressiva/genética , Antígenos CD/genética , Antígenos de Diferenciação Mielomonocítica/genética , Periodontite Crônica/genética , Estudo de Associação Genômica Ampla , Glicosiltransferases/genética , Lectinas/genética , Peptídeos Cíclicos/genética , alfa-Defensinas/genética , Estudos de Casos e Controles , Humanos , Polimorfismo de Nucleotídeo Único , Fatores de Risco
7.
J Bone Miner Res ; 32(3): 440-448, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27933643

RESUMO

Osteoblasts secrete matrix vesicles and proteins to bone surfaces, but the molecular mechanisms of this secretion system remain unclear. The present findings reveal the roles of important genes in osteoblasts involved in regulation of extracellular matrix secretion. We especially focused on "soluble N-ethylmaleimide-sensitive factor (NSF) attachment protein receptor" (SNARE) genes and identified notable Syntaxin 4a (Stx4a) expression on the basolateral side of the plasma membrane of osteoblasts. Furthermore, Stx4a overexpression was found to increase mineralization by osteoblastic cells, whereas Stx4a knockdown reduced levels of mineralization. Also, BMP-4 and IGF-1 induced the localization of Stx4a to the basolateral side of the cells. To examine the function of Stx4a in osteoblasts, we generated osteoblast-specific Stx4a conditional knockout mice, which demonstrated an osteopenic phenotype due to reduced matrix secretion. Bone mineral density, shown by peripheral quantitative computed tomography (pQCT), was reduced in the femur metaphyseal and diaphyseal regions of Stx4a osteoblast-specific deficient mice, whereas bone parameters, shown by micro-computed tomography (µCT) and bone histomorphometric analysis, were also decreased in trabecular bone. In addition, primary calvarial cells from those mice showed decreased mineralization and lower secretion of matrix vesicles. Our findings indicate that Stx4a plays a critical role in bone matrix production by osteoblasts. © 2016 American Society for Bone and Mineral Research.


Assuntos
Matriz Óssea/metabolismo , Vesículas Citoplasmáticas/metabolismo , Osteoblastos/metabolismo , Proteínas Qa-SNARE/metabolismo , Animais , Animais Recém-Nascidos , Densidade Óssea , Calcificação Fisiológica , Camundongos Knockout , Proteínas Qa-SNARE/genética , Crânio/citologia , Tíbia/diagnóstico por imagem , Microtomografia por Raio-X
8.
PLoS One ; 11(8): e0160765, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27509131

RESUMO

Periodontitis is an inflammatory disease causing loss of tooth-supporting periodontal tissue. Disease susceptibility to the rapidly progressive form of periodontitis, aggressive periodontitis (AgP), appears to be influenced by genetic risk factors. To identify these in a Japanese population, we performed whole exome sequencing of 41 unrelated generalized or localized AgP patients. We found that AgP is putatively associated with single nucleotide polymorphism (SNP) rs536714306 in the G-protein coupled receptor 126 gene, GPR126 [c.3086 G>A (p.Arg1029Gln)]. Since GPR126 activates the cAMP/PKA signaling pathway, we performed cAMP ELISA analysis of cAMP concentrations, and found that rs536714306 impaired the signal transactivation of GPR126. Moreover, transfection of human periodontal ligament (HPDL) cells with wild-type or mutant GPR126 containing rs536714306 showed that wild-type GPR126 significantly increased the mRNA expression of bone sialoprotein, osteopontin, and Runx2 genes, while mutant GPR126 had no effect on the expression of these calcification-related genes. The increase in expression of these genes was through the GPR126-induced increase of bone morphogenic protein-2, inhibitor of DNA binding (ID) 2, and ID4 expression. These data indicate that GPR126 might be important in maintaining the homeostasis of periodontal ligament tissues through regulating the cytodifferentiation of HPDL cells. The GPR126 SNP rs536714306 negatively influences this homeostasis, leading to the development of AgP, suggesting that it is a candidate genetic risk factor for AgP in the Japanese population.


Assuntos
Periodontite Agressiva/genética , Polimorfismo de Nucleotídeo Único , Receptores Acoplados a Proteínas G/genética , Adolescente , Adulto , Periodontite Agressiva/metabolismo , Povo Asiático/genética , Diferenciação Celular/genética , Exoma , Feminino , Estudo de Associação Genômica Ampla , Humanos , Masculino , Ligamento Periodontal/citologia , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/genética , Adulto Jovem
9.
Anticancer Drugs ; 26(3): 259-71, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25419631

RESUMO

Cyclic depsipeptides are polypeptides in which one or more amino acid is replaced by a hydroxy acid, resulting in the formation of at least one ester bond in the core ring structure. Many natural cyclic depsipeptides possessing intriguing structural and biological properties, including antitumor, antifungal, antiviral, antibacterial, anthelmintic, and anti-inflammatory activities, have been identified from fungi, plants, and marine organisms. In particular, the potent effects of cyclic depsipeptides on tumor cells have led to a number of clinical trials evaluating their potential as chemotherapeutic agents. Although many of the trials have not achieved the desired results, romidepsin (FK228), a bicyclic depsipeptide that inhibits histone deacetylase, has been shown to have clinical efficacy in patients with refractory cutaneous T-cell lymphoma and has received Food and Drug Administration approval for use in treatment. In this review, we discuss antitumor cyclic depsipeptides that have undergone clinical trials and focus on their structural features, mechanisms, potential applications in chemotherapy, and pharmacokinetic and toxicity data. The results of this study indicate that cyclic depsipeptides could be a rich source of new cancer therapeutics.


Assuntos
Antineoplásicos/farmacologia , Depsipeptídeos/farmacologia , Animais , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacocinética , Ensaios Clínicos como Assunto , Depsipeptídeos/efeitos adversos , Depsipeptídeos/farmacocinética , Inibidores de Histona Desacetilases/farmacologia , Humanos , Lactamas/efeitos adversos , Lactamas/farmacocinética , Lactamas/farmacologia , Lactonas/efeitos adversos , Lactonas/farmacocinética , Lactonas/farmacologia , Linfoma Cutâneo de Células T/tratamento farmacológico , Peptídeos Cíclicos
10.
Gene Expr Patterns ; 13(8): 372-6, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23872338

RESUMO

The Odd-skipped gene, first identified as a Drosophila pair-rule zinc-finger transcription factor, plays an important role in Drosophila development. The mammalian homolog, Odd-skipped related 2 (Osr2), regulates limb, tooth, and kidney development in mouse embryos. However, the detailed expression pattern of Osr2 during neonatal development remains unclear. In this study, we investigated Osr2 expression patterns in mouse neonatal and embryo tissues using qPCR and in situ hybridization methods. First, we examined the tissue distribution of Osr2 by qPCR, and found it to be highly expressed in the uterus and moderately in the testes, small intestine, and prostate. That expression was also found in eye, kidney, placenta, lung, thymus, lymph node, stomach, and skeletal muscle tissues, and in all embryonic stages. On the other hand, Osr2 was not expressed in brain, heart, liver, or spleen samples. Next, we examined the tissue localization of Osr2 using in situ hybridization. Osr2 was found in the craniofacial region on E13.5, with notable expression in dental germ mesenchyme as well as the renal corpuscle on E17.5. As for neonatal tissues, Osr2 was expressed in the dental papilla, dental follicle, Harderian gland, nasal bone, eyelid dermis, synovial joint, and tibial subcutis. Our findings suggest that Osr2 functions in reproductive system organs, such as the uterus, testes, prostate, placenta, and ovaries. Furthermore, based on its expression in kidney, Harderian gland, eyelid dermis, and tibial subcutis tissues, this transcription factor may be involved in hormone synthesis and function. Together, our results demonstrate the role of Osr2 in postnatal development and embryogenesis.


Assuntos
Expressão Gênica , Fatores de Transcrição/metabolismo , Animais , Osso e Ossos/metabolismo , Papila Dentária/metabolismo , Desenvolvimento Embrionário , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Intestino Delgado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos ICR , Mucosa Nasal/metabolismo , Especificidade de Órgãos , Placenta/metabolismo , Gravidez , Próstata/metabolismo , Testículo/metabolismo , Fatores de Transcrição/genética , Útero/metabolismo
11.
Development ; 138(24): 5369-78, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22110055

RESUMO

During development of the urogenital tract, fibroblast growth factor 8 (Fgf8) is expressed in mesonephric tubules, but its role in this tissue remains undefined. An evaluation of previously generated T-Cre-mediated Fgf8-deficient mice (T-Cre; Fgf8(flox/Δ2,3) mice), which lack Fgf8 expression in the mesoderm, revealed that the cranial region of the Wolffian duct degenerated prematurely and the cranial mesonephric tubules were missing. As a result, the epididymis, vas deferens and efferent ductules were largely absent in mutant mice. Rarb2-Cre was used to eliminate FGF8 from the mesonephric tubules but to allow expression in the adjacent somites. These mutants retained the cranial end of the Wolffian duct and formed the epididymis and vas deferens, but failed to elaborate the efferent ductules, indicating that Fgf8 expression by the mesonephric tubules is required specifically for the formation of the ductules. Ret knockout mice do not form the ureteric bud, a caudal outgrowth of the Wolffian duct and progenitor for the collecting duct network in the kidney, but they do develop the cranial end normally. This indicates that Fgf8, but not Ret, expression is essential to the outgrowth of the cranial mesonephric tubules from the Wolffian duct and to the development of major portions of the sex accessory tissues in the male reproductive tract. Mechanistically, FGF8 functions upstream of Lhx1 expression in forming the nephron, and analysis of Fgf8 mutants similarly shows deficient Lhx1 expression in the mesonephric tubules. These results demonstrate a multifocal requirement for FGF8 in establishing the male reproductive tract ducts and implicate Lhx1 signaling in tubule elongation.


Assuntos
Fator 8 de Crescimento de Fibroblasto/metabolismo , Genitália Masculina/crescimento & desenvolvimento , Ductos Mesonéfricos/crescimento & desenvolvimento , Animais , Regulação da Expressão Gênica no Desenvolvimento , Genitália Masculina/metabolismo , Proteínas com Homeodomínio LIM/metabolismo , Masculino , Mesoderma/crescimento & desenvolvimento , Mesoderma/metabolismo , Camundongos , Camundongos Knockout , Néfrons/crescimento & desenvolvimento , Néfrons/metabolismo , Proteínas Proto-Oncogênicas c-ret/metabolismo , Fatores de Transcrição/metabolismo , Sistema Urogenital/crescimento & desenvolvimento , Sistema Urogenital/metabolismo , Ductos Mesonéfricos/metabolismo
12.
Biochem Biophys Res Commun ; 414(1): 186-91, 2011 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-21946062

RESUMO

Korkormicin A belongs to a family of nature-produced cyclic depsipeptides. It has potent antitumor activity against both leukemia cell P388 and carcinoma cell M109. To further explore its potential as a cancer therapeutic, the mechanism of its antitumor activity was investigated. We found that korkormicin A can bind to DNA through intercalation. It also induces p53 phosphorylation, which leads to inhibition of p53 degradation and activation of p53-dependent transcription. Furthermore, korkormicin A preferentially induces apoptosis in transformed cells retaining wild type p53. As it has been shown that p53 usually induces apoptosis in transformed cells, but only growth arrest in untransformed cells, these results indicate that korkormicin A is a potential antitumor agent for cancers with wild type p53.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , DNA/farmacologia , Depsipeptídeos/farmacologia , Resistencia a Medicamentos Antineoplásicos , Substâncias Intercalantes/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Antineoplásicos/química , Linhagem Celular Tumoral , DNA/química , Depsipeptídeos/química , Humanos , Substâncias Intercalantes/química , Fosforilação/efeitos dos fármacos , Proteína Supressora de Tumor p53/genética , Ubiquitinação/efeitos dos fármacos
13.
J Biol Chem ; 285(19): 14585-93, 2010 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-20220130

RESUMO

Mammalian kidney development requires the functions of the Wilms tumor gene WT1 and the WNT/beta-catenin signaling pathway. Recent studies have shown that WT1 negatively regulates WNT/beta-catenin signaling, but the molecular mechanisms by which WT1 inhibits WNT/beta-catenin signaling are not completely understood. In this study, we identified a gene, CXXC5, which we have renamed WID (WT1-induced Inhibitor of Dishevelled), as a novel WT1 transcriptional target that negatively regulates WNT/beta-catenin signaling. WT1 activates WID transcription through the upstream enhancer region. In the developing kidney, Wid and Wt1 are coexpressed in podocytes of maturing nephrons. Structure-function analysis demonstrated that WID interacts with Dishevelled via its C-terminal CXXC zinc finger and Dishevelled binding domains and potently inhibits WNT/beta-catenin signaling in vitro and in vivo. WID is evolutionarily conserved, and ablation of wid in zebrafish embryos with antisense morpholino oligonucleotides perturbs embryonic kidney development. Taken together, our results demonstrate that the WT1 negatively regulates WNT/beta-catenin pathway via its target gene WID and further suggest a role for WID in nephrogenesis.


Assuntos
Proteínas de Transporte/metabolismo , Regulação Neoplásica da Expressão Gênica , Transdução de Sinais , Proteínas WT1/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteína Axina , Proteínas de Transporte/genética , Imunoprecipitação da Cromatina , Proteínas de Ligação a DNA , Proteínas Desgrenhadas , Regulação para Baixo , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Humanos , Immunoblotting , Imunoglobulina G/imunologia , Imunoprecipitação , Rim/citologia , Rim/metabolismo , Luciferases/metabolismo , Camundongos , Células NIH 3T3 , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/farmacologia , Coelhos , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição , Proteínas WT1/genética , Proteínas Wnt/genética , Peixe-Zebra , beta Catenina/genética
14.
Cancer Sci ; 100(1): 24-8, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19037995

RESUMO

The ubiquitin-proteasome system plays a critical role in controlling the level, activity and location of various cellular proteins. Significant progress has been made in investigating the molecular mechanisms of ubiquitination, particularly in understanding the structure of the ubiquitination machinery and identifying ubiquitin protein ligases, the primary specificity-determining enzymes. Therefore, it is now possible to target specific molecules involved in ubiquitination and proteasomal degradation to regulate many cellular processes such as signal transduction, proliferation and apoptosis. In particular, alterations in ubiquitination are observed in most, if not all, cancer cells. This is manifested by destabilization of tumor suppressors, such as p53, and overexpression of oncogenes such as c-Myc and c-Jun. In addition to the development and clinical validation of proteasome inhibitor, bortezomib, in myeloma therapy, recent studies have demonstrated that it is possible to develop inhibitors for specific ubiquitination and deubiquitination enzymes. With the help of structural studies, rational design and chemical synthesis, it is conceivable that we will be able to use 'druggable' inhibitors of the ubiquitin system to evaluate their effects in animal tumor models in the not-so-distant future.


Assuntos
Neoplasias/tratamento farmacológico , Inibidores de Proteases/uso terapêutico , Inibidores de Proteassoma , Ubiquitina/metabolismo , Enzima Desubiquitinante CYLD , Genes BRCA1 , Humanos , Neoplasias/metabolismo , Complexo de Endopeptidases do Proteassoma/fisiologia , Proteínas Quinases Associadas a Fase S/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/genética , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo
15.
Bioorg Med Chem ; 16(23): 10022-8, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18977148

RESUMO

Compounds that stabilize p53 could suppress tumors providing a additional tool to fight cancer. Mdm2, and the human ortholog, Hdm2 serve as ubiquitin E3 ligases and target p53 for ubiquitylation and degradation. Inhibition of Hdm2 stabilizes p53, inhibits cell proliferation and induces apoptosis. Using HTS to discover inhibitors, we identified three new alkaloids, isolissoclinotoxin B, diplamine B, and lissoclinidine B from Lissoclinum cf. badium. Lissoclinidine B inhibited ubiquitylation and degradation of p53, and selectively killed transformed cells harboring wild-type p53, suggesting this compound could be used to develop new treatments.


Assuntos
Alcaloides/farmacologia , Inibidores Enzimáticos/farmacologia , Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , Proteína Supressora de Tumor p53/metabolismo , Urocordados/química , Acridinas/química , Acridinas/isolamento & purificação , Acridinas/metabolismo , Alcaloides/química , Alcaloides/isolamento & purificação , Animais , Linhagem Celular Transformada , Inibidores Enzimáticos/química , Inibidores Enzimáticos/isolamento & purificação , Humanos , Concentração Inibidora 50 , Fenantrolinas/química , Fenantrolinas/isolamento & purificação , Fenantrolinas/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Ubiquitina/metabolismo
16.
Mol Cancer Ther ; 7(8): 2445-54, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18723490

RESUMO

The tumor suppressor protein p53 is a potent inducer of apoptosis in transformed cells. Hdm2 is an ubiquitin ligase (E3) that acts as a major regulator of p53 by promoting its ubiquitylation and proteasomal degradation. For this reason, inhibiting the E3 activity of Hdm2 has been proposed as a therapeutic approach for cancers expressing wild-type p53. We previously identified a family of small molecules (HLI98s, 7-nitro-10-aryl-5-deazaflavins) that inhibit the E3 activity of Hdm2, increase cellular p53, and selectively kill transformed cells expressing wild-type p53. However, issues of both potency and solubility in aqueous solution limit the utility of the HLI98s. Here, we report that a highly soluble derivative of the HLI98s, which has a 5-dimethylaminopropylamino side chain but lacks the 10-aryl group (HLI373), has greater potency than the HLI98s in stabilizing Hdm2 and p53, activating p53-dependent transcription, and inducing cell death. Furthermore, we show that HLI373 is effective in inducing apoptosis of several tumor cells lines that are sensitive to DNA-damaging agents. These results suggest that HLI373 could serve as a potential lead for developing cancer therapeutics based on inhibition of the ubiquitin ligase activity of Hdm2.


Assuntos
Aminoquinolinas/farmacologia , Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , Timina/análogos & derivados , Proteína Supressora de Tumor p53/metabolismo , Água/química , Apoptose/efeitos dos fármacos , Sequência de Bases , Linhagem Celular Transformada , Primers do DNA , Humanos , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Solubilidade , Timina/farmacologia , Proteína Supressora de Tumor p53/genética , Ubiquitina/metabolismo
17.
J Biomol Screen ; 13(3): 229-37, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18270365

RESUMO

High-throughput screening technologies have revolutionized the manner in which potential therapeutics are identified. Although they are the source of lead compounds for ~65% of anticancer and antimicrobial drugs approved by the Food and Drug Administration between 1981 and 2002, natural products have largely been excluded from modern screening programs. This is due, at least in part, to the inherent difficulties in testing complex extract mixtures, which often contain nuisance compounds, in modern bioassay systems. In this article, the authors present a novel electrochemiluminescent assay system for inhibition of MDM2 activity that is suitable for testing natural product extracts in high-throughput screening systems. The assay was used to screen more than 144,000 natural product extracts. The authors identified 1 natural product, sempervirine, that inhibited MDM2 auto-ubiquitination, MDM2-mediated p53 degradation, and led to accumulation of p53 in cells. Sempervirine preferentially induced apoptosis in transformed cells expressing wild-type p53, suggesting that it could be a potential lead for anticancer therapeutics.


Assuntos
Produtos Biológicos/farmacologia , Misturas Complexas/farmacologia , Avaliação Pré-Clínica de Medicamentos/métodos , Medições Luminescentes/métodos , Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , Animais , Bioensaio , Caspase 3/metabolismo , Morte Celular , Linhagem Celular Transformada , Camundongos , Poli(ADP-Ribose) Polimerases/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Alcaloides de Triptamina e Secologanina/química , Proteína Supressora de Tumor p53/metabolismo , Ubiquitinação/efeitos dos fármacos
18.
Cancer Res ; 67(19): 9472-81, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17909057

RESUMO

The conjugation of proteins with ubiquitin plays numerous regulatory roles through both proteasomal-dependent and nonproteasomal-dependent functions. Alterations in ubiquitylation are observed in a wide range of pathologic conditions, including numerous malignancies. For this reason, there is great interest in targeting the ubiquitin-proteasome system in cancer. Several classes of proteasome inhibitors, which block degradation of ubiquitylated proteins, are widely used in research, and one, Bortezomib, is now in clinical use. Despite the well-defined and central role of the ubiquitin-activating enzyme (E1), no cell permeable inhibitors of E1 have been identified. Such inhibitors should, in principle, block all functions of ubiquitylation. We now report 4[4-(5-nitro-furan-2-ylmethylene)-3,5-dioxo-pyrazolidin-1-yl]-benzoic acid ethyl ester (PYR-41) as the first such inhibitor. Unexpectedly, in addition to blocking ubiquitylation, PYR-41 increased total sumoylation in cells. The molecular basis for this is unknown; however, increased sumoylation was also observed in cells harboring temperature-sensitive E1. Functionally, PYR-41 attenuates cytokine-mediated nuclear factor-kappaB activation. This correlates with inhibition of nonproteasomal (Lys-63) ubiquitylation of TRAF6, which is essential to IkappaB kinase activation. PYR-41 also prevents the downstream ubiquitylation and proteasomal degradation of IkappaBalpha. Furthermore, PYR-41 inhibits degradation of p53 and activates the transcriptional activity of this tumor suppressor. Consistent with this, it differentially kills transformed p53-expressing cells. Thus, PYR-41 and related pyrazones provide proof of principle for the capacity to differentially kill transformed cells, suggesting the potential for E1 inhibitors as therapeutics in cancer. These inhibitors can also be valuable tools for studying ubiquitylation.


Assuntos
Benzoatos/farmacologia , Furanos/farmacologia , Pirazóis/farmacologia , Enzimas Ativadoras de Ubiquitina/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/metabolismo , Citocinas/metabolismo , Células HeLa , Humanos , Quinase I-kappa B/metabolismo , Células Jurkat , Camundongos , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma , Coelhos , Especificidade por Substrato , Proteína Supressora de Tumor p53/metabolismo , Ubiquitina/metabolismo , Enzimas Ativadoras de Ubiquitina/metabolismo
19.
Dev Biol ; 251(1): 142-56, 2002 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12413904

RESUMO

The Wnt antagonist Frzb-1 is expressed during limb skeletogenesis, but its roles in this complex multistep process are not fully understood. To address this issue, we determined Frzb-1 gene expression patterns during chick long bone development and carried out gain- and loss-of-function studies by misexpression of Frzb-1, Wnt-8 (a known Frzb-1 target), or different forms of the intracellular Wnt mediator LEF-1 in developing limbs and cultured chondrocytes. Frzb-1 expression was quite strong in mesenchymal prechondrogenic condensations and then characterized epiphyseal articular chondrocytes and prehypertrophic chondrocytes in growth plates. Virally driven Frzb-1 misexpression caused shortening of skeletal elements, joint fusion, and delayed chondrocyte maturation, with consequent inhibition of matrix mineralization, metalloprotease expression, and marrow/bone formation. In good agreement, misexpression of Frzb-1 or a dominant-negative form of LEF-1 in cultured chondrocytes maintained the cells at an immature stage. Instead, misexpression of Wnt-8 or a constitutively active LEF-1 strongly promoted chondrocyte maturation, hypertrophy, and calcification. Immunostaining revealed that the distribution of endogenous Wnt mediator beta-catenin changes dramatically in vivo and in vitro, from largely cytoplasmic in immature proliferating and prehypertrophic chondrocytes to nuclear in hypertrophic mineralizing chondrocytes. Misexpression of Frzb-1 prevented beta-catenin nuclear relocalization in chondrocytes in vivo or in vitro. The data demonstrate that Frzb-1 exerts a strong influence on limb skeletogenesis and is a powerful and direct modulator of chondrocyte maturation, phenotype, and function. Phases of skeletogenesis, such as terminal chondrocyte maturation and joint formation, appear to be particularly dependent on Wnt signaling and thus very sensitive to Frzb-1 antagonistic action.


Assuntos
Desenvolvimento Ósseo/fisiologia , Diferenciação Celular/fisiologia , Extremidades/embriologia , Glicoproteínas/fisiologia , Proteínas de Peixe-Zebra , Animais , Densidade Óssea/fisiologia , Calcificação Fisiológica/fisiologia , Embrião de Galinha , Condrócitos/citologia , Condrócitos/fisiologia , Extremidades/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/fisiologia , Transdução de Sinais , Proteínas Wnt
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA