Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Ageing Res Rev ; 82: 101766, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36283617

RESUMO

INTRODUCTION: The protein Klotho (KL) was first discovered in KL-deficient mice, which developed a syndrome similar to premature aging in humans. Since then, KL has been implicated in multiple molecular signaling pathways and diseases. KL has been shown to have anti-aging, healthspan and lifespan extending, cognitive enhancing, anti-oxidative, anti-inflammatory, and anti-tumor properties. KL levels decrease with age and in many diseases. Therefore, it has been of great interest to develop a KL-boosting or restoring drug, or to supplement endogenous Klotho with exogenous Klotho genetic material or recombinant Klotho protein, and to use KL levels in the body as a marker for the efficacy of such drugs and as a biomarker for the diagnosis and management of diseases. OBJECTIVE: The goal of this study was to provide a comprehensive review of KL levels across age groups in individuals who are healthy or have certain health conditions, using four sources: blood, cerebrospinal fluid, urine, and whole biopsy/necropsy tissue. By doing so, baseline KL levels can be identified across the lifespan, in the absence or presence of disease. In turn, these findings can be used to guide the development of future KL-based therapeutics and biomarkers, which will heavily rely on an individual's baseline KL range to be efficacious. METHODS: A total of 65 studies were collected primarily using the PubMed database. Research articles that were published up to April 2022 were included. Statistical analysis was conducted using RStudio. RESULTS: Mean and median blood KL levels in healthy individuals, mean blood KL levels in individuals with renal conditions, and mean blood KL levels in individuals with metabolic or endocrine conditions were shown to decrease with age. Similarly, CSF KL levels in patients with AD also declined compared with age-matched controls. CONCLUSIONS: The present study confirms the trend that KL levels in blood decrease with age in humans, among those who are healthy, and even further among those with renal and endocrine/metabolic illnesses. Further, by drawing this trend from multiple published works, we were able to provide a general idea of baseline KL ranges, specifically in blood in these populations. These data add to the current knowledge on normal KL levels in the body and how they change with time and in disease, and can potentially support efforts to create KL-based treatments and screening tools to better manage aging, renal, and metabolic/endocrine diseases.


Assuntos
Senilidade Prematura , Glucuronidase , Humanos , Camundongos , Animais , Proteínas Klotho , Envelhecimento/metabolismo , Longevidade/genética , Biomarcadores
2.
Cancers (Basel) ; 13(24)2021 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-34944918

RESUMO

Klotho is an anti-aging transmembrane protein, which can be shed and can function as a hormone. Accumulating data indicate that klotho is a tumor suppressor in a wide array of malignancies, and designate the subdomain KL1 as the active region of the protein towards this activity. We aimed to study the role of klotho as a tumor suppressor in pancreatic ductal adenocarcinoma (PDAC). Bioinformatics analyses of The Cancer Genome Atlas (TCGA) datasets revealed a correlation between the survival of PDAC patients, levels of klotho expression, and DNA methylation, and demonstrated a unique hypermethylation pattern of klotho in pancreatic tumors. The in vivo effects of klotho and KL1 were examined using three mouse models. Employing a novel genetic model, combining pancreatic klotho knockdown with a mutation in Kras, the lack of klotho contributed to PDAC generation and decreased mousece survival. In a xenograft model, administration of viral particles carrying sKL, a spliced klotho isoform containing the KL1 domain, inhibited pancreatic tumors. Lastly, treatment with soluble sKL prolonged survival of Pdx1-Cre; KrasG12D/+;Trp53R172H/+ (KPC) mice, a model known to recapitulate human PDAC. In conclusion, this study provides evidence that klotho is a tumor suppressor in PDAC. Furthermore, these data suggest that the levels of klotho expression and DNA methylation could have prognostic value in PDAC patients, and that administration of exogenous sKL may serve as a novel therapeutic strategy to treat PDAC.

3.
FASEB J ; 34(6): 7234-7246, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32347987

RESUMO

There is an unmet need for treatments for diseases associated with aging. The antiaging, life-extending, and cognition-enhancing protein Klotho is neuroprotective due to its anti-inflammatory, antioxidative, and pro-myelinating effects. In addition, Klotho is also a tumor suppressor and has beneficial roles in multiple organs. Klotho is downregulated as part of the aging process. Thus, upregulating Klotho in the brain may lead to novel therapeutics to people suffering or at risk for neurodegenerative diseases such as Alzheimer's, Parkinson's, and amyotrophic lateral sclerosis, and demyelinating diseases such as multiple sclerosis. We attempted to upregulate Klotho for its beneficial effects in the brain and elsewhere. Here, we describe a method to specifically activate Klotho gene expression. To accomplish this task, we designed zinc finger proteins (ZFPs) targeting within -300 bps of the human Klotho promoter. We designed the ZPF constructs either de novo from modular building blocks, or modified sequences from the natural endogenous Egr1 transcription factor backbone structure. Egr1 is known to upregulate Klotho expression. We tested the transcriptional activation effects of these ZFPs in a dual luciferase coincidence reporter system under the control of 4-kb promoter of human Klotho in stable HEK293 cells and in HK-2 cells that express Klotho protein endogenously. We found that the best ZFPs are the de novo designed ones targeting -250 bps of Klotho promoter and one of the Egr1-binding sites. We further enhanced Klotho's activation using p65-Rta transcriptional activation domains in addition to VP64. These upregulation approaches could be useful for studying Klotho's protective effects and designing Klotho boosting therapeutics for future in vivo experiments.


Assuntos
Proteína 1 de Resposta de Crescimento Precoce/genética , Glucuronidase/genética , Regiões Promotoras Genéticas/genética , Dedos de Zinco/genética , Envelhecimento/genética , Sítios de Ligação/genética , Encéfalo/metabolismo , Linhagem Celular , Cognição/fisiologia , Expressão Gênica/genética , Células HEK293 , Humanos , Proteínas Klotho , Luciferases/genética , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Ativação Transcricional/genética , Regulação para Cima/genética
4.
PLoS One ; 15(1): e0226382, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31929539

RESUMO

Klotho is an age-extending, cognition-enhancing protein found to be down-regulated in aged mammals when age-related diseases start to appear. Low levels of Klotho occur in neurodegenerative diseases, kidney disease and many cancers. Many normal and pathologic processes involve the proteolytic shedding of membrane proteins. Transmembrane (TM) Klotho contains two homologous domains, KL1 and KL2 with homology to glycosidases. After shedding by ADAM 10 and 17, a shed Klotho isoform is released into serum and urine by the kidney, and into the CSF by the choroid plexus. We previously reported that human Klotho contains two major cleavage sites. However, the exact cleavage site responsible for the cleavage between the KL1 and KL2 domains remains unknown for the human Klotho, and both sites are unknown for mouse Klotho. In this study, we aimed to identify the cleavage sites leading to the shed forms of human and mouse Klotho. Mutations in the region close to the TM domain of mouse Klotho result in the reduced shedding of the 130 kD (KL1+KL2) and 70 kD (KL1) fragments, suggesting that the cleavage site lies within the mutated region. We further identified the cleavage sites responsible for the cleavage between KL1 and KL2 of human and mouse Klotho. Moreover, mutated Klotho proteins have similar subcellular localization patterns as wild type Klotho. Finally, in an FGF23 functional assay, all Klotho mutants with a nine amino acid deletion can also function as an FGFR1 co-receptor for FGF23 signaling, however, the signaling activity was greatly reduced. The study provides new and important information on Klotho shedding, and paves the way for studies aimed to distinguish between the distinct roles of the various isoforms of Klotho.


Assuntos
Glucuronidase/metabolismo , Proteína ADAM10/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/metabolismo , Glucuronidase/química , Glucuronidase/genética , Células HEK293 , Humanos , Proteínas Klotho , Camundongos , Microscopia de Fluorescência , Mutagênese , Domínios Proteicos , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Alinhamento de Sequência , Transdução de Sinais
5.
PLoS One ; 14(3): e0213321, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30830941

RESUMO

BACKGROUND: Fibroblast growth factor 23 is an emerging vascular biomarker, recently associated with cerebral small vessel disease and poor cognition in patients on dialysis. It also interacts with klotho, an anti-aging and cognition enhancing protein. OBJECTIVE: To determine if circulating Fibroblast growth factor 23 (FGF23) is associated with new-onset cognitive outcomes in a community-based cohort of cognitively healthy adults with long-term follow-up. METHODS: We measured serum FGF23 levels in 1537 [53% women, mean age 68.7 (SD 5.7)] dementia-free Framingham Offspring participants at their 7th quadrennial examination (1998-2001), and followed these participants for the development of clinical all-cause dementia and Alzheimer's disease (AD). Secondary outcomes included MRI-based structural brain measures, and neurocognitive test performance at exam 7. RESULTS: During a median (Q1, Q3) 12-year (7.0, 13.3) follow up, 122 (7.9%) participants developed dementia, of whom 91 (5.9%) had AD. Proportional-hazards regression analysis, adjusted for age, sex, education, systolic blood pressure, antihypertensive medication, prevalent cardiovascular disease, diabetes mellitus, smoking status and apoE ε4 carrier status, revealed that higher serum FGF23 levels were associated with an increased risk of incident dementia and AD (Hazard ratio [HR] per 1 standard deviation increment in inverse transformed FGF23 level 1.25, 95% CI 1.02-1.53, and 1.32, 95% CI 1.04-1.69, respectively). There was no significant interaction according to presence/absence of significant renal impairment (eGFR <30 versus ≥30ml/min) and risk of dementia (based on 1537; p = 0.97). CONCLUSIONS: Higher circulating FGF23 is associated with an increased risk of dementia, suggesting that FGF23-related biological pathways may play a role in the development of dementia.


Assuntos
Doença de Alzheimer/diagnóstico , Biomarcadores/sangue , Demência/diagnóstico , Fatores de Crescimento de Fibroblastos/sangue , Idoso , Doença de Alzheimer/sangue , Doença de Alzheimer/epidemiologia , Pressão Sanguínea , Demência/sangue , Demência/epidemiologia , Feminino , Fator de Crescimento de Fibroblastos 23 , Humanos , Incidência , Estudos Longitudinais , Masculino , Estudos Prospectivos , Fatores de Risco , Estados Unidos/epidemiologia
6.
J Alzheimers Dis ; 67(3): 1089-1106, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30776010

RESUMO

Alzheimer's disease (AD) is characterized by the accumulation of neurotoxic amyloid-ß (Aß) peptides consisting of 39-43 amino acids, proteolytically derived fragments of the amyloid-ß protein precursor (AßPP), and the accumulation of the hyperphosphorylated microtubule-associated protein tau. Inhibiting Aß production may reduce neurodegeneration and cognitive dysfunction associated with AD. We have previously used an AßPP-firefly luciferase enzyme complementation assay to conduct a high throughput screen of a compound library for inhibitors of AßPP dimerization, and identified a compound that reduces Aß levels. In the present study, we have identified an analog, compound Y10, which also reduced Aß. Initial kinase profiling assays identified the receptor tyrosine kinase cKit as a putative Y10 target. To elucidate the precise mechanism involved, AßPP phosphorylation was examined by IP-western blotting. We found that Y10 inhibits cKit phosphorylation and increases AßPP phosphorylation mainly on tyrosine residue Y743, according to AßPP751 numbering. A known cKit inhibitor and siRNA specific to cKit were also found to increase AßPP phosphorylation and lower Aß levels. We also investigated a cKit downstream signaling molecule, the Shp2 phosphatase, and found that known Shp2 inhibitors and siRNA specific to Shp2 also increase AßPP phosphorylation, suggesting that the cKit signaling pathway is also involved in AßPP phosphorylation and Aß production. We further found that inhibitors of both cKit and Shp2 enhance AßPP surface localization. Thus, regulation of AßPP phosphorylation by small molecules should be considered as a novel therapeutic intervention for AD.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Transdução de Sinais/efeitos dos fármacos , Peptídeos beta-Amiloides/efeitos dos fármacos , Precursor de Proteína beta-Amiloide/efeitos dos fármacos , Western Blotting , Eletroforese em Gel de Poliacrilamida , Ensaio de Imunoadsorção Enzimática , Células HEK293 , Humanos
7.
Geroscience ; 40(1): 31-47, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29357021

RESUMO

Mammalian aging is associated with decline in cognitive functions. Studies searching for a cause of cognitive aging initially focused on neuronal loss but quantitative investigations of rat, monkey, and human brain using stereology demonstrated that in normal aging, unlike in neurodegenerative disease, neurons are not lost. Instead, electron microscopic and MRI studies in normal aging monkeys revealed age-related damage to myelin sheaths, loss of axons, and reduction in white matter volume which correlates with cognitive impairments. However, little is known about the cause of myelin defects or associated axon loss. The present study investigates the effect of age on signaling pathways between oligodendroglia and neurons using a custom PCR array to assess the expression of 87 genes of interest in cortical gray matter and white matter from the inferior parietal lobe (IPL) of normal rhesus monkeys ranging in age from 4.2 to 30.4 years old. From this array data, five target genes of interest were selected for further analysis to confirm gene expression and measure protein expression. The most interesting target gene identified is brain-derived neurotrophic factor (BDNF), which was the only gene that was altered at both mRNA and protein levels. In gray matter, BDNF mRNA was decreased. While the level of the mature form of the protein was unchanged, there was a specific decrease in the precursor form of BDNF. These alterations in the BDNF in gray matter could contribute to the vulnerability and loss of the axons with age.


Assuntos
Envelhecimento/fisiologia , Fator Neurotrófico Derivado do Encéfalo/genética , Encéfalo/patologia , Disfunção Cognitiva/metabolismo , Animais , Biópsia por Agulha , Encéfalo/metabolismo , Cognição/fisiologia , Disfunção Cognitiva/genética , Feminino , Estudos de Associação Genética/métodos , Imuno-Histoquímica , Macaca mulatta , Masculino , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real/métodos , Valores de Referência , Substância Branca/metabolismo
8.
J Mol Neurosci ; 64(2): 175-184, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29352444

RESUMO

Multiple lines of evidence show that the anti-aging and cognition-enhancing protein Klotho fosters neuronal survival, increases the anti-oxidative stress defense, and promotes remyelination of demyelinated axons. Thus, upregulation of the Klotho gene can potentially alleviate the symptoms and/or prevent the progression of age-associated neurodegenerative diseases such as Alzheimer's disease and demyelinating diseases such as multiple sclerosis. Here we used a CRISPR-dCas9 complex to investigate single-guide RNA (sgRNA) targeting the Klotho promoter region for efficient transcriptional activation of the Klotho gene. We tested the sgRNAs within the - 1 to - 300 bp of the Klotho promoter region and identified two sgRNAs that can effectively enhance Klotho gene transcription. We examined the transcriptional activation of the Klotho gene using three different systems: a Firefly luciferase (FLuc) and NanoLuc luciferase (NLuc) coincidence reporter system, a NLuc knock-in in Klotho 3'-UTR using CRISPR genomic editing, and two human cell lines: neuronal SY5Y cells and kidney HK-2 cells that express Klotho endogenously. The two sgRNAs enhanced Klotho expression at both the gene and protein levels. Our results show the feasibility of gene therapy for targeting Klotho using CRISPR technology. Enhancing Klotho levels has a therapeutic potential for increasing cognition and treating age-associated neurodegenerative, demyelinating and other diseases, such as chronic kidney disease and cancer.


Assuntos
Sistemas CRISPR-Cas , Glucuronidase/genética , Ativação Transcricional , Edição de Genes/métodos , Glucuronidase/metabolismo , Células HEK293 , Humanos , Proteínas Klotho , Regulação para Cima
9.
J Mol Neurosci ; 55(1): 76-90, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24907942

RESUMO

Klotho functions as an aging suppressor, which, in mice, extends lifespan when overexpressed and accelerates development of aging-like phenotypes when disrupted. Klotho is mainly expressed in brain and kidney and is secreted into the serum and CSF. We have previously shown that Klotho is reduced in brains of old monkeys, rats, and mice. We further reported the ability of Klotho to enhance oligodendrocyte differentiation and myelination. Here, we examined the signaling pathways induced by Klotho in MO3.13, a human oligodendrocytic hybrid cell line. We show that exogenous Klotho affects the ERK and Akt signaling pathways, decreases the proliferative abilities and enhances differentiation of MO3.13 cells. Furthermore, microarray analysis of Klotho-treated MO3.13 cells reveals a massive change in gene expression with 80 % of the differentially expressed genes being downregulated. Using gene set enrichment analysis, we predicted potential transcription factors involved in regulating Klotho-treated MO3.13 cells and found that these cells are highly enriched in the gene sets, that are similarly observed in cancer, cardiovascular disease, stress, aging, and hormone-related chemical and genetic perturbations. Since Klotho is downregulated in all brain tumors tested to date, enhancing Klotho has therapeutic potential for treating brain and other malignancies.


Assuntos
Glucuronidase/farmacologia , Neurogênese , Oligodendroglia/metabolismo , Animais , Linhagem Celular , Proliferação de Células , Humanos , Proteínas Klotho , Sistema de Sinalização das MAP Quinases , Camundongos , Oligodendroglia/citologia , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/fisiologia , Proteínas Recombinantes/farmacologia , Fatores de Transcrição/metabolismo
10.
Age (Dordr) ; 36(1): 141-9, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23818104

RESUMO

Klotho is an anti-aging protein with direct effects on life-span in mice. Klotho functions to regulate pathways classically associated with longevity including insulin/IGF1 and Wnt signaling. Decreased Klotho protein expression is observed throughout the body during the normal aging process. While increased methylation of the Klotho promoter is reported, other epigenetic mechanisms could contribute to age-related downregulation of Klotho expression, including microRNA-mediated regulation. Following in silico identification of potential microRNA binding sites within the Klotho 3' untranslated region, reporter assays reveal regulation by microRNA-339, microRNA-556, and, to a lesser extent, microRNA-10 and microRNA-199. MicroRNA-339 and microRNA-556 were further found to directly decrease Klotho protein expression indicating that, if upregulated in aging tissue, these microRNA could play a role in age-related downregulation of Klotho messenger RNA. These microRNAs are differentially regulated in cancer cells compared to normal cells and may imply a role for microRNA-mediated regulation of Klotho in cancer.


Assuntos
Envelhecimento/fisiologia , Glucuronidase/genética , Longevidade/genética , MicroRNAs/genética , Células Cultivadas , Genes Reporter , Glucuronidase/metabolismo , Humanos , Técnicas In Vitro , Fator de Crescimento Insulin-Like I/metabolismo , Proteínas Klotho , Luciferases , Neoplasias/genética , Regiões Promotoras Genéticas , RNA , Via de Sinalização Wnt/fisiologia
11.
Biochem J ; 441(1): 453-61, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21939436

RESUMO

The absence of Klotho (KL) from mice causes the development of disorders associated with human aging and decreased longevity, whereas increased expression prolongs lifespan. With age, KL protein levels decrease, and keeping levels consistent may promote healthier aging and be disease-modifying. Using the KL promoter to drive expression of luciferase, we conducted a high-throughput screen to identify compounds that activate KL transcription. Hits were identified as compounds that elevated luciferase expression at least 30%. Following validation for dose-dependent activation and lack of cytotoxicity, hit compounds were evaluated further in vitro by incubation with opossum kidney and Z310 rat choroid plexus cells, which express KL endogenously. All compounds elevated KL protein compared with control. To determine whether increased protein resulted in an in vitro functional change, we assayed FGF23 (fibroblast growth factor 23) signalling. Compounds G-I augmented ERK (extracellular-signal-regulated kinase) phosphorylation in FGFR (fibroblast growth factor receptor)-transfected cells, whereas co-transfection with KL siRNA (small interfering RNA) blocked the effect. These compounds will be useful tools to allow insight into the mechanisms of KL regulation. Further optimization will provide pharmacological tools for in vivo studies of KL.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Glucuronidase/metabolismo , Envelhecimento/fisiologia , Animais , Linhagem Celular , Clonagem Molecular , Ensaios de Seleção de Medicamentos Antitumorais , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/farmacologia , Regulação da Expressão Gênica/fisiologia , Glucuronidase/genética , Rim/citologia , Proteínas Klotho , Camundongos , Gambás , Ratos
12.
J Neurochem ; 100(2): 458-67, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17241160

RESUMO

Considerable evidence indicates that the amyloid-beta (Abeta) peptide, a proteolytic fragment of the amyloid precursor protein, is the pathogenic agent in Alzheimer's disease (AD). A number of proteases have been reported as capable of degrading Abeta, among them: neprilysin, insulin-degrading enzyme, endothelin-converting enzyme-1 and -2, angiotensin-converting enzyme and plasmin. These proteases, originating from a variety of cell types, degrade Abeta of various conformational states and in different cellular locations. We report here the isolation of a serine protease from serum-free conditioned medium of human neuroblastoma cells. Tandem mass spectrometry (MS/MS)-based sequencing of the isolated protein identified acyl peptide hydrolase (APH; EC3.4.19.1) as the active peptidase. APH is one of four members of the prolyl oligopeptidase family of serine proteases expressed in a variety of cells and tissues, including erythrocytes, liver and brain, but its precise biological activity is unknown. Here, we describe the identification of APH as an Abeta-degrading enzyme, and we show that the degradation of Abeta by APH isolated from transfected cells is inhibited by APH-specific inhibitors, as well as by synthetic Abeta peptide. In addition, we cloned APH from human brain and from neuroblastoma cells. Most importantly, our results indicate that APH expression in AD brain is lower than in age-matched controls.


Assuntos
Aciltransferases/metabolismo , Peptídeos beta-Amiloides/metabolismo , Meios de Cultivo Condicionados/química , Serina/metabolismo , Aciltransferases/genética , Animais , Autorradiografia/métodos , Linhagem Celular , Chlorocebus aethiops , Inibidores da Colinesterase , Meios de Cultivo Condicionados/farmacologia , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Humanos , Isoflurofato/farmacocinética , Mutação/fisiologia , Neuroblastoma/enzimologia , Neuroblastoma/patologia , Ligação Proteica/efeitos dos fármacos , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Serina/genética , Espectrometria de Massas em Tandem/métodos , Transfecção , Trítio/farmacocinética
13.
Immunity ; 18(3): 429-40, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12648459

RESUMO

Most antigenic peptides presented on MHC class I molecules are generated by proteasomes during protein breakdown. It is unknown whether these peptides are protected from destruction by cytosolic peptidases. In cytosolic extracts, most antigenic peptides are degraded by the metalloendopeptidase, thimet oligopeptidase (TOP). We therefore examined whether TOP destroys antigenic peptides in vivo. When TOP was overexpressed in cells, class I presentation of antigenic peptides was reduced. In contrast, TOP overexpression didn't reduce presentation of peptides generated in the endoplasmic reticulum or endosomes. Conversely, preventing TOP expression with siRNA enhanced presentation of antigenic peptides. TOP therefore plays an important role in vivo in degrading peptides released by proteasomes and is a significant factor limiting the extent of antigen presentation.


Assuntos
Apresentação de Antígeno/fisiologia , Antígenos/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Metaloendopeptidases/metabolismo , Peptídeos/imunologia , Peptídeos/metabolismo , Animais , Células COS , Membrana Celular/imunologia , Membrana Celular/metabolismo , Cisteína Endopeptidases/metabolismo , Citosol/imunologia , Citosol/metabolismo , Retículo Endoplasmático/imunologia , Retículo Endoplasmático/metabolismo , Expressão Gênica , Células HeLa , Humanos , Metaloendopeptidases/genética , Camundongos , Complexos Multienzimáticos/metabolismo , Complexo de Endopeptidases do Proteassoma
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA