Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Neuroscience ; 540: 38-47, 2024 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-38242280

RESUMO

Secretory clusterin (sCLU) plays an important role in the research progress of nervous system diseases. However, the physiological function of sCLU in Parkinson's disease (PD) are unclear. The purpose of this study was to examine the effects of sCLU-mediated autophagy on cell survival and apoptosis inhibition in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of PD. We found that MPTP administration induced prolonged pole-climbing time, shortened traction time and rotarod time, significantly decreased TH protein expression in the SN tissue of mice. In contrast, sCLU -treated mice took less time to climb the pole and had an extended traction time and rotating rod time. Meanwhile, sCLU intervention induced increased expression of the TH protein in the SN of mice. These results indicated that sCLU intervention could reduce the loss of dopamine neurons in the SN area and alleviate dyskinesia in mice. Furthermore, MPTP led to suppressed viability, enhanced apoptosis, an increased Bax/Bcl-2 ratio, and cleaved caspase-3 in the SN of mice, and these effects were abrogated by sCLU intervention. In addition, MPTP increased the levels of P62 protein, decreased Beclin1 protein, decreased the ratio of LC3B-II/LC3B-I, and decreased the numbers of autophagosomes and autophagolysosomes in the SN tissues of mice. These effects were also abrogated by sCLU intervention. Activation of PI3K/AKT/mTOR signaling with MPTP inhibited autophagy in the SN of MPTP mice; however, sCLU treatment activated autophagy in MPTP-induced PD mice by inhibiting PI3K/AKT/mTOR signaling. These data indicated that sCLU treatment had a neuroprotective effect in an MPTP-induced model of PD.


Assuntos
Fármacos Neuroprotetores , Doença de Parkinson , Animais , Camundongos , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Apoptose , Autofagia , Clusterina/metabolismo , Clusterina/farmacologia , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/metabolismo , Doença de Parkinson/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo
2.
BMB Rep ; 57(3): 149-154, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37817436

RESUMO

The stomach has emerged as a crucial endocrine organ in the regulation of feeding since the discovery of ghrelin. Gut-derived hormones, such as ghrelin and cholecystokinin, can act through the vagus nerve. We previously reported the satiety effect of hypothalamic clusterin, but the impact of peripheral clusterin remains unknown. In this study, we administered clusterin intraperitoneally to mice and observed its ability to suppress fasting-driven food intake. Interestingly, we found its synergism with cholecystokinin and antagonism with ghrelin. These effects were accompanied by increased c-fos immunoreactivity in nucleus tractus solitarius, area postrema, and hypothalamic paraventricular nucleus. Notably, truncal vagotomy abolished this response. The stomach expressed clusterin at high levels among the organs, and gastric clusterin was detected in specific enteroendocrine cells and the submucosal plexus. Gastric clusterin expression decreased after fasting but recovered after 2 hours of refeeding. Furthermore, we confirmed that stomachspecific overexpression of clusterin reduced food intake after overnight fasting. These results suggest that gastric clusterin may function as a gut-derived peptide involved in the regulation of feeding through the gut-brain axis. [BMB Reports 2024; 57(3): 149-154].


Assuntos
Ingestão de Alimentos , Grelina , Camundongos , Animais , Grelina/farmacologia , Ingestão de Alimentos/fisiologia , Clusterina/farmacologia , Colecistocinina/farmacologia , Estômago , Comportamento Alimentar
3.
Epilepsy Behav ; 138: 109016, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36473302

RESUMO

The developing infant brain has a different response mechanism and repair potential for injury than the adult brain. There is an urgent need for new anticonvulsants to effectively control neonatal seizures while minimizing the drug's toxic damage to the developing brain. Leptin protects neuronal plasma membrane integrity, while it has clinical advantages in terms of anticonvulsant properties as well. This study aimed to evaluate the effect of immediate leptin treatment on the serum concentration of clusterin and vascular endothelial growth factor (VEGF), neuronal plasma membrane integrity-related proteins, and the neurobehavioral phenotypes following neonatal seizures. Leptin was injected i.p at a dose of 4 mg/kg 1 hour after daily 30 minutes prolonged seizures for consecutive 10 days. The serum biomarkers (clusterin and VEGF), and brain protein expression of ATF-4/GRP78/autophagy axis were measured by enzyme-linked immunosorbent assay and western blot in the acute phase (24 hours after the last seizures), respectively. Behavioral and histopathological phenotypes and seizure threshold were conducted from P23 to P34, respectively. There were rapid elevation of serum VEGF and clusterin as well as upregulated protein expression of ATF-4, GRP78, Beclin-1, and LC3 in the cerebral cortex and hippocampus following a neonatal seizure, which was restored by immediate treatment with leptin after seizures. In addition, leptin improved seizure-induced impaired neuropsychological, and cognitive functioning. Furthermore, leptin succeeded in ameliorating markers of neuronal excitability, including seizure threshold and hippocampal mossy fiber sprouting. In conclusion, this study verified that immediate treatment with leptin after neonatal seizures restored both rapid elevation of serum clusterin as well as upregulated protein expression of ATF-4/GRP78/autophagy axis in the cerebral cortex and hippocampus, which contributes to the recovery of neurological function.


Assuntos
Epilepsia , Fator A de Crescimento do Endotélio Vascular , Animais , Ratos , Fator A de Crescimento do Endotélio Vascular/farmacologia , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Proteínas de Choque Térmico/farmacologia , Ratos Sprague-Dawley , Leptina , Clusterina/genética , Clusterina/metabolismo , Clusterina/farmacologia , Chaperona BiP do Retículo Endoplasmático , Convulsões , Encéfalo , Hipocampo/patologia , Epilepsia/metabolismo , Fenótipo , Estresse Oxidativo
4.
Biol Trace Elem Res ; 201(8): 3755-3773, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36394793

RESUMO

It is now widely accepted that anti-cancer medications are most effective when administered in combination. Zinc is an essential micronutrient whilst berberine is a well-known natural phytochemical, both having multiple molecular mechanisms of action. The present study aimed to determine the combinatorial effect of zinc and berberine on the human adenocarcinoma HT-29 cancer cell line. The anti-proliferative activity of berberine and zinc was determined by cell viability and colony-forming assays. The combination index and drug reduction index values of zinc and berberine co-treatments were estimated by suitable software. Flow cytometry was used to analyse cell cycle distribution and Annexin V/PI staining. The expression of apoptosis and zinc signalling markers were analysed by RT-qPCR and immunoblot analysis. Berberine decreased the viability of colon cancer cells in a dose-dependent manner whilst zinc alone had no significant influence on it. However, zinc and berberine co-treatment resulted in a synergistic anti-cancer action which was demonstrated by G2/M phase arrest of cell growth at a lower dose of berberine. Annexin V assay demonstrated that the synergistic impact of zinc and berberine boosted the number of apoptotic cells. Gene expression analysis at both transcriptional and translational levels showed the upregulation of apoptotic (caspase-3 and caspase-8) and a zinc-sensing receptor (GPR39) gene with concomitant downregulation of anti-apoptotic genes like proliferating cell nuclear antigen (PCNA) and clusterin. Our findings showed that the combination of zinc and berberine has synergistic anti-cancer efficacy and thus could be used as a potential chemopreventive option for colon cancer.


Assuntos
Berberina , Neoplasias do Colo , Neoplasias Colorretais , Humanos , Berberina/farmacologia , Berberina/uso terapêutico , Zinco/farmacologia , Clusterina/farmacologia , Anexina A5/farmacologia , Linhagem Celular Tumoral , Apoptose , Proliferação de Células , Neoplasias do Colo/tratamento farmacológico , Neoplasias Colorretais/tratamento farmacológico , Receptores Acoplados a Proteínas G
5.
J Neuroinflammation ; 19(1): 311, 2022 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-36550510

RESUMO

Preclinical studies have recently evaluated the impact of low-dose brain radiation therapy (LD-RT) in animal models of Alzheimer's disease (AD) showing anti-amyloid and anti-inflammatory effects of this treatment. Its effectiveness varied, however, depending on the LD-RT protocol used and the stage when the treatment was applied. In this study, we aimed to evaluate the therapeutic potential of 10 Gy delivered in five daily fractions of 2 Gy (a protocol previously shown to induce an improvement of cognitive performances) in 9-month-old TgF344-AD rats, modeling at a pre-symptomatic stage of the disease. We showed that at an early stage, LD-RT was able to lower levels of the 18-kDa translocator protein (TSPO)-mediated neuroinflammation to normal ranges in addition to the secreted CLUSTERIN, another inflammatory protein also involved in Aß aggregation. In addition, we demonstrated that LD-RT reduces all amyloid forms (~ - 60 to - 80%, P < 0.01; soluble and aggregated forms of Aß40, Aß42, and Aßoligomers). Interestingly, we showed for the first time that sAPPα levels were improved by the treatment, showing a higher activation of the non-amyloidogenic pathway, that could favor neuronal survival. The current evidence confirms the capacity of LD-RT to successfully modulate two pathological hallmarks of AD, namely amyloid and neuroinflammation, when applied before symptoms onset.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Ratos , Animais , Peptídeos beta-Amiloides/metabolismo , Clusterina/metabolismo , Clusterina/farmacologia , Doenças Neuroinflamatórias , Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Amiloide/metabolismo , Modelos Animais de Doenças , Proteínas de Transporte/metabolismo , Receptores de GABA-A
6.
J Cell Physiol ; 237(7): 3012-3029, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35567755

RESUMO

This study provides comprehensive mechanistic evidence for the role of clusterin, a stress-response secretory chaperone protein, in the modulation of intraocular pressure (IOP) by regulating the trabecular meshwork (TM) actin cytoskeleton and the extracellular matrix (ECM). The pathological stressors on TM known to elevate IOP significantly lowered clusterin protein levels indicating stress-related clusterin function loss. Small interfering RNA-mediated clusterin loss in human TM cells in vitro induced actin polymerization and stabilization via protein kinase D1, serine/threonine-protein kinase N2 (PRK2), and LIM kinase 1 (LIMK1), and the recruitment and activation of adhesome proteins including paxillin, vinculin, and integrin αV and ß5. A complete loss of clusterin as seen in clusterin knockout mice (Clu-/- ) led to significant IOP elevation at postnatal Day 70. Contrarily, constitutive clusterin expression using adenovirus (AdCLU) in HTM cells resulted in the loss of actin polymerization via decreased PRK2, and LIMK1 and negative regulation of integrin αV and ß5. Furthermore, we found that AdCLU treatment in HTM cells significantly decreased the ECM protein expression and distribution by significantly increasing matrix metalloprotease 2 (MMP2) activity and lowering the levels of pro-fibrotic proteins such as transforming growth factor-ß2 (TGFß2), thrombospondin-1 (TSP-1), and plasminogen activator inhibitor-1 (PAI-1). Finally, we found that HTM cells supplemented with recombinant human clusterin attenuated the pro-fibrotic effects of TGFß2. For the first time this study demonstrates the importance of clusterin in the regulation of TM actin cytoskeleton - ECM interactions and the maintenance of IOP, thus making clusterin an interesting target to reverse elevated IOP.


Assuntos
Pressão Intraocular , Malha Trabecular , Actinas/metabolismo , Animais , Células Cultivadas , Clusterina/genética , Clusterina/metabolismo , Clusterina/farmacologia , Matriz Extracelular/metabolismo , Humanos , Integrina alfaV/metabolismo , Integrina alfaV/farmacologia , Quinases Lim/metabolismo , Camundongos , Polimerização , Fator de Crescimento Transformador beta2/farmacologia
7.
Ann Surg Oncol ; 29(8): 4937-4946, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35397747

RESUMO

BACKGROUND: The outcome of pancreatic ductal adenocarcinoma (PDAC) is unsatisfactory, and the identification of novel therapeutic targets is urgently needed. Clinical studies on the antisense oligonucleotide that targets clusterin (CLU) expression have been conducted and have shown efficacy in other cancers. We aimed to investigate the effects of CLU in PDAC and the underlying mechanisms with a view to the clinical application of existing drugs. METHODS: We knocked down CLU in PDAC cells and evaluated changes in cell proliferation. To elucidate the mechanism responsible for these changes, we performed western blot analysis, cell cycle assay, and senescence-associated ß-galactosidase (SA-ß-gal) staining. To evaluate the clinical significance of CLU, immunohistochemistry was performed, and CLU expression was analyzed in specimens resected from PDAC patients not treated with preoperative chemotherapy. RESULTS: Knockdown of CLU significantly decreased cell proliferation and did not induce apoptosis, but did induce cellular senescence by increasing the percentage of G1-phase and SA-ß-gal staining-positive cells. A marker of DNA damage such as γH2AX and factors related to cellular senescence, such as p21 and the senescence-associated secretory phenotype, were upregulated by knockdown of CLU. CLU expression in resected PDAC specimens was located in the cytoplasm of tumor cells and revealed significantly better recurrence-free survival and overall survival in the CLU-low group than in the CLU-high group. CONCLUSIONS: We identified that CLU inhibition leads to cellular senescence in PDAC. Our findings suggest that CLU is a novel therapeutic target that contributes to the prognosis of PDAC by inducing cellular senescence.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Carcinoma Ductal Pancreático/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Senescência Celular , Clusterina/genética , Clusterina/farmacologia , Humanos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas
8.
Biochim Biophys Acta Mol Basis Dis ; 1868(7): 166384, 2022 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-35292360

RESUMO

Clusterin is a heterodimeric glycoprotein (α- and ß-chain), which has been described as an extracellular molecular chaperone. In humans, clusterin is an amyloid-associated protein, co-localizing with fibrillar deposits in several amyloidoses, including Alzheimer's disease. To clarify its potential implication in amyloid formation, we located aggregation-prone regions within the sequence of clusterin α-chain, via computational methods. We had peptide-analogues, which correspond to each of these regions, chemically synthesized and experimentally demonstrated that all of them can form amyloid-like fibrils. We also provide evidence that the same peptide-analogues can inhibit amyloid-ß fibril formation, potentially making them appropriate drug candidates for Alzheimer's disease. At the same time, our findings hint that the respective aggregation-prone clusterin regions may be implicated in the molecular mechanism in which clusterin inhibits amyloid formation. Furthermore, we suggest that molecular chaperones with amyloidogenic properties might have a role in the regulation of amyloid formation, essentially acting as functional amyloids.


Assuntos
Doença de Alzheimer , Amiloidose , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Amiloide/metabolismo , Amiloidose/tratamento farmacológico , Clusterina/química , Clusterina/metabolismo , Clusterina/farmacologia , Glicoproteínas , Humanos
9.
Folia Biol (Praha) ; 67(5-6): 183-190, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35439851

RESUMO

Apolipoprotein J (clusterin) is a component of high-density lipoproteins, the high level of which is reversely correlated with the risk of coronary heart disease. In addition, it exerts anti-inflammatory and anti-apoptotic effects on endothelial cells and inhibits smooth muscle cell migration and proliferation, indicating that it may play a protective role in cardiovascular disease. However, the exact mechanisms by which this occurs remain unclear. This study aimed to clarify these underlying protective mechanisms by researching the inhibitory effects of apolipoprotein J via the NOD-like receptor protein 3 pathway on the inflammation induced by cholesterol crystals in THP­1 macrophages. In culture, THP-1 macrophages were infected with adenoviral vectors containing apolipoprotein J genes and subsequently treated with cholesterol crystals. The inflammatory cytokines interleukin­1ß, interleukin 18 and tumour necrosis factor α were quantitatively measured with ELISA kits. NOD-like receptor protein 3, cysteinyl aspartate specific proteinase 1 and interleukin 1ß were evaluated by Western blot and PCR analysis. As a result, apolipoprotein J expression was found to remarkably decrease the levels of inflammatory cytokines, including tumour necrosis factor α, interleukin 18 and interleukin 1ß, secreted by THP­1 macrophages. It was also found capable of inhibiting the levels of NOD-like receptor protein 3, cysteinyl aspartate-specific proteinase 1 and interleukin 1ß both at the protein and mRNA levels. In the current study, we revealed that over-expression of apolipoprotein J attenuated the inflammation induced by cholesterol crystals through inhibition of the NOD-like receptor protein 3 inflammasome pathway.


Assuntos
Inflamassomos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Ácido Aspártico/metabolismo , Ácido Aspártico/farmacologia , Colesterol/metabolismo , Clusterina/metabolismo , Clusterina/farmacologia , Citocinas/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Humanos , Inflamassomos/metabolismo , Inflamassomos/farmacologia , Inflamação/patologia , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Peptídeo Hidrolases/metabolismo , Peptídeo Hidrolases/farmacologia , Fator de Necrose Tumoral alfa/metabolismo
10.
Endocrinol Metab (Seoul) ; 35(4): 943-953, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33261311

RESUMO

BACKGROUND: There is a great need to discover factors that could protect pancreatic ß-cells from apoptosis and thus prevent diabetes mellitus. Clusterin (CLU), a chaperone protein, plays an important role in cell protection in numerous cells and is involved in various cellular mechanisms, including autophagy. In the present study, we investigated the protective role of CLU through autophagy regulation in pancreatic ß-cells. METHODS: To identify the protective role of CLU, mouse insulinoma 6 (MIN6) cells were incubated with CLU and/or free fatty acid (FFA) palmitate, and cellular apoptosis and autophagy were examined. RESULTS: Treatment with CLU remarkably upregulated microtubule-associated protein 1-light chain 3 (LC3)-II conversion in a doseand time-dependent manner with a significant increase in the autophagy-related 3 (Atg3) gene expression level, which is a mediator of LC3-II conversion. Moreover, co-immunoprecipitation and fluorescence microscopy experiments showed that the molecular interaction of LC3 with Atg3 and p62 was markedly increased by CLU. Stimulation of LC3-II conversion by CLU persisted in lipotoxic conditions, and FFA-induced apoptosis and dysfunction were simultaneously improved by CLU treatment. Finally, inhibition of LC3-II conversion by Atg3 gene knockdown markedly attenuated the cytoprotective effect of CLU. CONCLUSION: Taken together, these findings suggest that CLU protects pancreatic ß-cells against lipotoxicity-induced apoptosis via autophagy stimulation mediated by facilitating LC3-II conversion. Thus, CLU has therapeutic effects on FFA-induced pancreatic ß-cell dysfunction.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Clusterina/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Camundongos , Proteínas Associadas aos Microtúbulos/efeitos dos fármacos , Palmitatos/toxicidade , Substâncias Protetoras/farmacologia , Regulação para Cima
11.
Osteoarthritis Cartilage ; 28(5): 685-697, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31981738

RESUMO

OBJECTIVE: Innate immune response and particularly terminal complement complex (TCC) deposition are thought to be involved in the pathogenesis of posttraumatic osteoarthritis. However, the possible role of TCC in regulated cell death as well as chondrocyte hypertrophy and senescence has not been unraveled so far and was first addressed using an ex vivo human cartilage trauma-model. DESIGN: Cartilage explants were subjected to blunt impact (0.59 J) and exposed to human serum (HS) and cartilage homogenate (HG) with or without different potential therapeutics: RIPK1-inhibitor Necrostatin-1 (Nec), caspase-inhibitor zVAD, antioxidant N-acetyl cysteine (NAC) and TCC-inhibitors aurintricarboxylic acid (ATA) and clusterin (CLU). Cell death and hypertrophy/senescence-associated markers were evaluated on mRNA and protein level. RESULTS: Addition of HS resulted in significantly enhanced TCC deposition on chondrocytes and decrease of cell viability after trauma. This effect was potentiated by HG and was associated with expression of RIPK3, MLKL and CASP8. Cytotoxicity of HS could be prevented by heat-inactivation or specific inhibitors, whereby combination of Nec and zVAD as well as ATA exhibited highest cell protection. Moreover, HS+HG exposition enhanced the gene expression of CXCL1, IL-8, RUNX2 and VEGFA as well as secretion of IL-6 after cartilage trauma. CONCLUSIONS: Our findings imply crucial involvement of the complement system and primarily TCC in regulated cell death and phenotypic changes of chondrocytes after cartilage trauma. Inhibition of TCC formation or downstream signaling largely modified serum-induced pathophysiologic effects and might therefore represent a therapeutic target to maintain the survival and chondrogenic character of cartilage cells.


Assuntos
Morte Celular/genética , Condrócitos/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento/genética , Hipertrofia/genética , Osteoartrite/genética , Ferimentos não Penetrantes/genética , Acetilcisteína/farmacologia , Idoso , Idoso de 80 Anos ou mais , Ácido Aurintricarboxílico/farmacologia , Cartilagem Articular/citologia , Morte Celular/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Senescência Celular/genética , Condrócitos/efeitos dos fármacos , Condrócitos/patologia , Clusterina/farmacologia , Complexo de Ataque à Membrana do Sistema Complemento/antagonistas & inibidores , Complexo de Ataque à Membrana do Sistema Complemento/efeitos dos fármacos , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Inibidores Enzimáticos/farmacologia , Feminino , Sequestradores de Radicais Livres/farmacologia , Humanos , Imidazóis/farmacologia , Imunidade Inata/genética , Indóis/farmacologia , Masculino , Pessoa de Meia-Idade , Oligopeptídeos/farmacologia , Osteoartrite/etiologia , Osteoartrite/metabolismo , Osteoartrite/patologia , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ferimentos não Penetrantes/complicações , Ferimentos não Penetrantes/metabolismo
12.
Artigo em Inglês | MEDLINE | ID: mdl-31672573

RESUMO

Mimetic peptides are promising therapeutic agents for atherosclerosis prevention. A 10-residue class G* peptide from apolipoprotein J (apoJ), namely, D-[113-122]apoJ, possesses anti-inflammatory and anti-atherogenic properties. This prompted us to determine its effect on the aggregation process of low-density lipoprotein (LDL) particles, an early event in the development of atherosclerosis. LDL particles with and without [113-122]apoJ peptide were incubated at 37 °C with sphingomyelinase (SMase) or were left to aggregate spontaneously at room temperature. The aggregation process was analyzed by size-exclusion chromatography (SEC), native gradient gel electrophoresis (GGE), absorbance at 405 nm, dynamic light scattering (DLS), and transmission electronic microscopy (TEM). In addition, circular dichroism was used to determine changes in the secondary structure of apoB, and SDS-PAGE was performed to assess apoB degradation. At an equimolar ratio of [113-122]apoJ peptide to apoB-100, [113-122]apoJ inhibited both SMase-induced or spontaneous LDL aggregation. All methods showed that [113-122]apoJ retarded the progression of SMase-induced LDL aggregation at long incubation times. No effect of [113-122]apoJ on apoB secondary structure was observed. Binding experiments showed that [113-122]apoJ presents low affinity for native LDL but binds readily to LDL during the first stages of aggregation. Laurdan fluorescence experiments showed that mild aggregation of LDL resulted in looser lipid packaging, which was partially prevented by D-[113-122]apoJ. These results demonstrate that [113-122]apoJ peptide prevents SMase-induced LDL aggregation at an equimolar ratio and opens the possibility for the use of this peptide as a therapeutic tool.


Assuntos
Clusterina/farmacologia , Lipoproteínas LDL/metabolismo , Fragmentos de Peptídeos/farmacologia , Agregados Proteicos/efeitos dos fármacos , Esfingomielinas/metabolismo , Aterosclerose/metabolismo , Aterosclerose/prevenção & controle , Clusterina/química , Clusterina/uso terapêutico , Voluntários Saudáveis , Humanos , Lipoproteínas LDL/sangue , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/uso terapêutico , Esfingomielina Fosfodiesterase/metabolismo , Esfingomielinas/sangue
13.
Diabetes Care ; 42(3): 466-475, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30659075

RESUMO

OBJECTIVE: Components of the adipose tissue (AT) extracellular matrix (ECM) are recently discovered contributors to obesity-related cardiometabolic disease. We identified increased adipocyte expression of ECM-related clusterin (apolipoprotein J) in obese versus lean women by microarray. Our objective was to determine 1) whether subcutaneous AT adipocyte (SAd) clusterin and serum clusterin are associated with insulin resistance (IR) and known markers of cardiometabolic risk and 2) how clusterin may contribute to increased risk. RESEARCH DESIGN AND METHODS: We validated increased clusterin expression in adipocytes from a separate group of 18 lean and 54 obese individuals. The relationship of clusterin gene expression and plasma clusterin with IR, cardiovascular biomarkers, and risk of cardiovascular disease (CVD) was then determined. Further investigations in human cultured cells and in aged LDLR-/- mice prone to development of obesity-associated complications were performed. RESULTS: SAd clusterin correlated with IR, multiple CVD biomarkers, and CVD risk, independent of traditional risk factors. Circulating human clusterin exhibited similar associations. In human adipocytes, palmitate enhanced clusterin secretion, and in human hepatocytes, clusterin attenuated insulin signaling and APOA1 expression and stimulated hepatic gluconeogenesis. LRP2 (megalin), a clusterin receptor, highly expressed in liver, mediated these effects, which were inhibited by LRP2 siRNA. In response to Western diet feeding, an increase in adipocyte clusterin expression was associated with a progressive increase in liver fat, steatohepatitis, and fibrosis in aged LDLR-/- mice. CONCLUSIONS: Adipocyte-derived clusterin is a novel ECM-related protein linking cardiometabolic disease and obesity through its actions in the liver.


Assuntos
Adipócitos/metabolismo , Clusterina/fisiologia , Resistência à Insulina/genética , Insulina/metabolismo , Fígado/efeitos dos fármacos , Adipócitos/efeitos dos fármacos , Tecido Adiposo/metabolismo , Adulto , Animais , Biomarcadores/metabolismo , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/metabolismo , Células Cultivadas , Clusterina/genética , Clusterina/farmacologia , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Feminino , Células Hep G2 , Humanos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Obesidade/sangue , Obesidade/genética , Obesidade/metabolismo , Receptores de LDL/genética , Fatores de Risco , Gordura Subcutânea/metabolismo
14.
Cell Physiol Biochem ; 45(3): 1003-1012, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29428944

RESUMO

BACKGROUND/AIMS: Ischemia-reperfusion (I/R) injury is an unavoidable event occurring during heart transplantation and is a key factor in graft failure and the long-term survival rate of recipients. Therefore, there is an urgent need for the development of new therapies to prevent I/R injury. Clusterin is a hetero-dimeric glycoprotein with an antiapoptotic function. In this study, we investigated whether clusterin was cardioprotective in heart transplantation against I/R injury using an in vivo rat model and an in vitro cell culture system, and examined the underlying mechanisms of I/R injury. METHODS: Heart grafts from wild-type C57BL/6 mice were preserved in UW solution (control) or UW solution containing recombinant human apolipoprotein-J (hr clusterin) for 24 h. The preserved hearts were implanted into recipient mice of the same strain as the donors for 72 h, and the heart grafts were then taken for histopathological and gene expression analyses. An in vitro ischemia reperfusion model using H9C2 cells or H9C2/clusterin cDNA cells was constructed. The expression of clusterin, p65, Bax, Bcl-xL, IL-1ß, and TNF-α protein and mRNA in heart tissue and H9C2 cells was detected by western blot, reverse transcription-polymerase chain reaction (RT-PCR), and quantitative RT-PCR assays; IL-1ß and TNF-α protein was detected by enzyme-linked immunosorbent assays; NF-kB activity was detected by an electrophoretic mobility shift assay; cell apoptosis was detected by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling and flow cytometric analyses. RESULTS: Cold I/R caused severe morphologic myocardial injury to heart grafts from wild-type C57BL/6 mice, whereas grafts from hr clusterin preservation showed less damage, as demonstrated by decreased cell apoptosis/death, decreased neutrophil infiltration, and the preservation of the normal structure of the heart. Clusterin reduced the expression of p65, pre-inflammatory IL-1ß, and TNF-α, and the pro-apoptotic gene Bax, while it enhanced the expression of the anti-apoptotic gene Bcl-xL in vitro and in vivo. Clusterin inhibited cell apoptosis/death and reduced pre-inflammatory. CONCLUSION: Clusterin is a promising target for preventing cold I/R injury in heart transplantation. This study also shows that the resultant protective effects of clusterin are mediated by NF-κB signaling and Bax/Bcl-xL expression.


Assuntos
Clusterina/farmacologia , Transplante de Coração , Coração/efeitos dos fármacos , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteína X Associada a bcl-2/genética , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Clusterina/genética , Clusterina/metabolismo , Regulação da Expressão Gênica , Interleucina-1beta/análise , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miocárdio/patologia , Preservação de Órgãos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/prevenção & controle , Transdução de Sinais , Fator de Necrose Tumoral alfa/análise , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Proteína X Associada a bcl-2/metabolismo
15.
BMC Neurosci ; 19(1): 2, 2018 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-29370749

RESUMO

BACKGROUND: Alzheimer's disease (AD) is characterized by the deposition of amyloid-ß (Aß) in brain parenchyma and cerebral blood vessels as cerebral amyloid angiopathy (CAA). Clusterin, a chaperon protein associated with Aß aggregation, toxicity and transport through blood-brain barrier, may play a key role in the development of AD. Recently, clusterin peptide D-[113-122] was shown to mimic clusterin's function and exerted therapeutic effect in atherosclerosis. In this study, we investigated whether this clusterin peptide also affected (Aß) deposition in AD transgenic mouse. RESULTS: Using a micropump, synthetic peptide 113-122 of clusterin protein (20 µg/200 µl) was infused into the lateral ventricle of 8-month 5 × FAD transgenic mouse model (Tg6799), for 2 weeks. Water-maze testing showed an improved cognitive function of the Tg6799 mice treated with clusterin. Immunocytochemistry and quantitative analysis revealed that intraventricular (icv) administration of clusterin peptide in Tg6799 mouse reduced Aß plaques as well the severity of cerebral amyloid angiopathy. Enzyme-linked immunosorbent assay demonstrated a decreased in the soluble levels of Aß (Aß40 and Aß42) in the brain. Western-blot revealed an increased level of LRP-2 after clusterin peptide treatment. CONCLUSION: These results suggest that icv infusion of clusterin peptide D-[113-122] offers a promising therapeutic approach to reduce Aß deposition as well as CAA. The LRP2-mediated clearance system might be involved in the mechanism of these effects.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Barreira Hematoencefálica/efeitos dos fármacos , Angiopatia Amiloide Cerebral/tratamento farmacológico , Clusterina/farmacologia , Cognição/efeitos dos fármacos , Peptídeos beta-Amiloides/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Clusterina/administração & dosagem , Modelos Animais de Doenças , Infusões Intraventriculares , Camundongos Transgênicos
16.
Artigo em Inglês | MEDLINE | ID: mdl-28159830

RESUMO

Normal tau homeostasis is achieved when the synthesis, processing, and degradation of the protein is balanced. Together, the pathways that regulate tau homeostasis ensure that the protein is at the proper levels and that its posttranslational modifications and subcellular localization are appropriately controlled. These pathways include the enzymes responsible for posttranslational modifications, those systems that regulate mRNA splicing, and the molecular chaperones that control tau turnover and its binding to microtubules. In tauopathies, this delicate balance is disturbed. Tau becomes abnormally modified by posttranslational modification, it loses affinity for microtubules, and it accumulates in proteotoxic aggregates. How and why does this imbalance occur? In this review, we discuss how molecular chaperones and other components of the protein homeostasis (e.g., proteostasis) network normally govern tau quality control. We also discuss how aging might reduce the capacity of these systems and how tau mutations might further affect this balance. Finally, we discuss how small-molecule inhibitors are being used to probe and perturb the tau quality-control systems, playing a particularly prominent role in revealing the logic of tau homeostasis. As such, there is now interest in developing these chemical probes into therapeutics, with the goal of restoring normal tau homeostasis to treat disease.


Assuntos
Proteínas de Choque Térmico/metabolismo , Proteostase , Tauopatias/metabolismo , Proteínas tau/metabolismo , Animais , Clusterina/metabolismo , Clusterina/farmacologia , Proteínas de Choque Térmico/antagonistas & inibidores , Proteínas de Choque Térmico/farmacologia , Humanos , Mutação de Sentido Incorreto , Ligação Proteica , Tauopatias/tratamento farmacológico , Proteínas tau/antagonistas & inibidores
17.
Cell Rep ; 21(11): 3310-3316, 2017 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-29241555

RESUMO

One potential therapeutic strategy for Alzheimer's disease (AD) is to use antibodies that bind to small soluble protein aggregates to reduce their toxic effects. However, these therapies are rarely tested in human CSF before clinical trials because of the lack of sensitive methods that enable the measurement of aggregate-induced toxicity at low concentrations. We have developed highly sensitive single vesicle and single-cell-based assays that detect the Ca2+ influx caused by the CSF of individuals affected with AD and healthy controls, and we have found comparable effects for both types of samples. We also show that an extracellular chaperone clusterin; a nanobody specific to the amyloid-ß peptide (Aß); and bapineuzumab, a humanized monoclonal antibody raised against Aß, could all reduce the Ca2+ influx caused by synthetic Aß oligomers but are less effective in CSF. These assays could be used to characterize potential therapeutic agents in CSF before clinical trials.


Assuntos
Peptídeos beta-Amiloides/antagonistas & inibidores , Bioensaio , Cálcio/metabolismo , Líquido Cefalorraquidiano/química , Vesículas Citoplasmáticas/efeitos dos fármacos , Fragmentos de Peptídeos/antagonistas & inibidores , Agregados Proteicos/efeitos dos fármacos , Idoso , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Sequência de Aminoácidos , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/imunologia , Animais , Anticorpos Monoclonais Humanizados/farmacologia , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Clusterina/farmacologia , Meios de Cultura/farmacologia , Vesículas Citoplasmáticas/metabolismo , Feminino , Humanos , Transporte de Íons/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/imunologia , Cultura Primária de Células , Ratos , Anticorpos de Domínio Único/farmacologia
18.
Acta Neuropathol Commun ; 5(1): 81, 2017 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-29115989

RESUMO

It is now widely accepted in the field that the normally secreted chaperone clusterin is redirected to the cytosol during endoplasmic reticulum (ER) stress, although the physiological function(s) of this physical relocation remain unknown. We have examined in this study whether or not increased expression of clusterin is able to protect neuronal cells against intracellular protein aggregation and cytotoxicity, characteristics that are strongly implicated in a range of neurodegenerative diseases. We used the amyotrophic lateral sclerosis-associated protein TDP-43 as a primary model to investigate the effects of clusterin on protein aggregation and neurotoxicity in complementary in vitro, neuronal cell and Drosophila systems. We have shown that clusterin directly interacts with TDP-43 in vitro and potently inhibits its aggregation, and observed that in ER stressed neuronal cells, clusterin co-localized with TDP-43 and specifically reduced the numbers of cytoplasmic inclusions. We further showed that the expression of TDP-43 in transgenic Drosophila neurons induced ER stress and that co-expression of clusterin resulted in a dramatic clearance of mislocalized TDP-43 from motor neuron axons, partially rescued locomotor activity and significantly extended lifespan. We also showed that in Drosophila photoreceptor cells, clusterin co-expression gave ER stress-dependent protection against proteotoxicity arising from both Huntingtin-Q128 and mutant (R406W) human tau. We therefore conclude that increased expression of clusterin can provide an important defense against intracellular proteotoxicity under conditions that mimic specific features of neurodegenerative disease.


Assuntos
Clusterina/metabolismo , Clusterina/farmacologia , Proteínas de Ligação a DNA/metabolismo , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/metabolismo , Síndromes Neurotóxicas/tratamento farmacológico , Animais , Animais Geneticamente Modificados , Linhagem Celular Tumoral , Clusterina/genética , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Estresse do Retículo Endoplasmático/genética , Olho/metabolismo , Olho/ultraestrutura , Hemolinfa/citologia , Humanos , Líquido Intracelular/efeitos dos fármacos , Líquido Intracelular/metabolismo , Larva , Atividade Motora/genética , Atividade Motora/fisiologia , Neurônios Motores/ultraestrutura , Neuroblastoma/patologia , Síndromes Neurotóxicas/genética , Síndromes Neurotóxicas/patologia , Agregados Proteicos/efeitos dos fármacos , Agregados Proteicos/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transfecção
19.
PLoS One ; 12(11): e0188191, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29155887

RESUMO

Aß-Immunotherapy has long been studied in the treatment of Alzheimer's disease (AD), but not how other molecules involved in the disease can affect antibody performance. We previously designed an antibody fragment, scFv-h3D6, and showed that it precludes Aß-induced cytotoxicity by withdrawing Aß oligomers from the amyloid pathway towards a non-toxic, worm-like pathway. ScFv-h3D6 was effective at the behavioral, cellular, and molecular levels in the 3xTg-AD mouse model. Because scFv-h3D6 treatment restored apolipoprotein E (apoE) and J (apoJ) concentrations to non-pathological values, and Aß internalization by glial cells was found to be decreased in the presence of these apolipoproteins, we now aimed to test the influence of scFv-h3D6 on Aß aggregation and cellular uptake by primary human astrocytes in the presence of therapeutic apoE and apoJ mimetic peptides (MPs). Firstly, we demonstrated by CD and FTIR that the molecules used in this work were well folded. Next, interactions between apoE or apoJ-MP, scFv-h3D6 and Aß were studied by CD. The conformational change induced by the interaction of Aß with apoE-MP was much bigger than the induced with apoJ-MP, in line with the observed formation of protective worm-like fibrils by the scFv-h3D6/Aß complex in the presence of apoJ-MP, but not of apoE-MP. ScFv-h3D6, apoJ-MP, and apoE-MP to a different extent reduced Aß uptake by astrocytes, and apoE-MP partially interfered with the dramatic reduction by scFv-h3D6 while apoJ-MP had no effect on scFv-h3D6 action. As sustained Aß uptake by astrocytes may impair their normal functions, and ultimately neuronal viability, this work shows another beneficence of scFv-h3D6 treatment, which is not further improved by the use of apoE or apoJ mimetic peptides.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Astrócitos/efeitos dos fármacos , Fragmentos de Peptídeos/metabolismo , Peptídeos/farmacologia , Agregados Proteicos/efeitos dos fármacos , Anticorpos de Cadeia Única/farmacologia , Adulto , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/genética , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Apolipoproteínas E/farmacologia , Astrócitos/metabolismo , Astrócitos/patologia , Clonagem Molecular , Clusterina/genética , Clusterina/metabolismo , Clusterina/farmacologia , Endocitose/efeitos dos fármacos , Escherichia coli/genética , Escherichia coli/metabolismo , Feminino , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Mimetismo Molecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Peptídeos/genética , Peptídeos/metabolismo , Cultura Primária de Células , Dobramento de Proteína , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Anticorpos de Cadeia Única/biossíntese , Anticorpos de Cadeia Única/genética
20.
PLoS One ; 12(8): e0182389, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28767729

RESUMO

Retinitis Pigmentosa (RP) begins with the death of rod photoreceptors and is slowly followed by a gradual loss of cones and a rearrangement of the remaining retinal neurons. Clusterin is a chaperone protein that protects cells and is involved in various pathophysiological stresses, including retinal degeneration. Using a well-established transgenic rat model of RP (rhodopsin S334ter), we investigated the effects of clusterin on rod photoreceptor survival. To investigate the role of clusterin in S334ter-line3 retinas, Voronoi analysis and immunohistochemistry were used to evaluate the geometry of rod distribution. Additionally, immunoblot analysis, Bax activation, STAT3 and Akt phosphorylation were used to evaluate the pathway involved in rod cell protection. In this study, clusterin (10µg/ml) intravitreal treatment produced robust preservation of rod photoreceptors in S334ter-line3 retina. The mean number of rods in 1mm2 was significantly greater in clusterin injected RP retinas (postnatal (P) 30, P45, P60, & P75) than in age-matched saline injected RP retinas (P<0.01). Clusterin activated Akt, STAT3 and significantly reduced Bax activity; in addition to inducing phosphorylated STAT3 in Müller cells, which suggests it may indirectly acts on photoreceptors. Thus, clusterin treatment may interferes with mechanisms leading to rod death by suppressing cell death through activation of Akt and STAT3, followed by Bax suppression. Novel insights into the pathway of how clusterin promotes the rod cell survival suggest this treatment may be a potential therapeutic strategy to slow progression of vision loss in human RP.


Assuntos
Clusterina/administração & dosagem , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/efeitos dos fármacos , Retinose Pigmentar/tratamento farmacológico , Fator de Transcrição STAT3/metabolismo , Proteína X Associada a bcl-2/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Clusterina/farmacologia , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Injeções Intravítreas , Fosforilação , Ratos , Células Fotorreceptoras Retinianas Bastonetes/patologia , Retinose Pigmentar/genética , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA