Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
2.
Cell ; 161(2): 291-306, 2015 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-25860611

RESUMO

Cholesterol is dynamically transported among organelles, which is essential for multiple cellular functions. However, the mechanism underlying intracellular cholesterol transport has remained largely unknown. We established an amphotericin B-based assay enabling a genome-wide shRNA screen for delayed LDL-cholesterol transport and identified 341 hits with particular enrichment of peroxisome genes, suggesting a previously unappreciated pathway for cholesterol transport. We show dynamic membrane contacts between peroxisome and lysosome, which are mediated by lysosomal Synaptotagmin VII binding to the lipid PI(4,5)P2 on peroxisomal membrane. LDL-cholesterol enhances such contacts, and cholesterol is transported from lysosome to peroxisome. Disruption of critical peroxisome genes leads to cholesterol accumulation in lysosome. Together, these findings reveal an unexpected role of peroxisome in intracellular cholesterol transport. We further demonstrate massive cholesterol accumulation in human patient cells and mouse model of peroxisomal disorders, suggesting a contribution of abnormal cholesterol accumulation to these diseases.


Assuntos
Colesterol/metabolismo , Lisossomos/metabolismo , Peroxissomos/metabolismo , RNA Interferente Pequeno/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Adrenoleucodistrofia/metabolismo , Anfotericina B/farmacologia , Animais , Transporte Biológico , Estudo de Associação Genômica Ampla , Humanos , Camundongos , Transtornos Peroxissômicos/metabolismo , Transtornos Peroxissômicos/patologia , Fosfatidilinositol 4,5-Difosfato/metabolismo , Sinaptotagminas/metabolismo , Peixe-Zebra
3.
Mol Cell ; 66(1): 154-162.e10, 2017 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-28344083

RESUMO

Hedgehog (Hh) has been known as the only cholesterol-modified morphogen playing pivotal roles in development and tumorigenesis. A major unsolved question is how Hh signaling regulates the activity of Smoothened (SMO). Here, we performed an unbiased biochemical screen and identified that SMO was covalently modified by cholesterol on the Asp95 (D95) residue through an ester bond. This modification was inhibited by Patched-1 (Ptch1) but enhanced by Hh. The SMO(D95N) mutation, which could not be cholesterol modified, was refractory to Hh-stimulated ciliary localization and failed to activate downstream signaling. Furthermore, homozygous SmoD99N/D99N (the equivalent residue in mouse) knockin mice were embryonic lethal with severe cardiac defects, phenocopying the Smo-/- mice. Together, the results of our study suggest that Hh signaling transduces to SMO through modulating its cholesterylation and provides a therapeutic opportunity to treat Hh-pathway-related cancers by targeting SMO cholesterylation.


Assuntos
Colesterol/metabolismo , Proteínas Hedgehog/metabolismo , Transdução de Sinais , Receptor Smoothened/metabolismo , Animais , Células CHO , Cílios/metabolismo , Cricetulus , Regulação da Expressão Gênica no Desenvolvimento , Predisposição Genética para Doença , Células HEK293 , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/metabolismo , Proteínas Hedgehog/genética , Humanos , Camundongos , Camundongos Transgênicos , Mutação , Células NIH 3T3 , Receptor Patched-1/genética , Receptor Patched-1/metabolismo , Fenótipo , Processamento de Proteína Pós-Traducional , Interferência de RNA , Receptor Smoothened/genética , Transfecção
4.
Nature ; 531(7596): 651-5, 2016 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-26982734

RESUMO

CD8(+) T cells have a central role in antitumour immunity, but their activity is suppressed in the tumour microenvironment. Reactivating the cytotoxicity of CD8(+) T cells is of great clinical interest in cancer immunotherapy. Here we report a new mechanism by which the antitumour response of mouse CD8(+) T cells can be potentiated by modulating cholesterol metabolism. Inhibiting cholesterol esterification in T cells by genetic ablation or pharmacological inhibition of ACAT1, a key cholesterol esterification enzyme, led to potentiated effector function and enhanced proliferation of CD8(+) but not CD4(+) T cells. This is due to the increase in the plasma membrane cholesterol level of CD8(+) T cells, which causes enhanced T-cell receptor clustering and signalling as well as more efficient formation of the immunological synapse. ACAT1-deficient CD8(+) T cells were better than wild-type CD8(+) T cells at controlling melanoma growth and metastasis in mice. We used the ACAT inhibitor avasimibe, which was previously tested in clinical trials for treating atherosclerosis and showed a good human safety profile, to treat melanoma in mice and observed a good antitumour effect. A combined therapy of avasimibe plus an anti-PD-1 antibody showed better efficacy than monotherapies in controlling tumour progression. ACAT1, an established target for atherosclerosis, is therefore also a potential target for cancer immunotherapy.


Assuntos
Acetatos/farmacologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Colesterol/metabolismo , Imunoterapia/métodos , Melanoma/tratamento farmacológico , Melanoma/imunologia , Ácidos Sulfônicos/farmacologia , Acetamidas , Acetatos/uso terapêutico , Acetil-CoA C-Acetiltransferase/antagonistas & inibidores , Acetil-CoA C-Acetiltransferase/deficiência , Acetil-CoA C-Acetiltransferase/genética , Acetil-CoA C-Acetiltransferase/metabolismo , Animais , Aterosclerose/tratamento farmacológico , Linfócitos T CD8-Positivos/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Esterificação/efeitos dos fármacos , Feminino , Sinapses Imunológicas/efeitos dos fármacos , Sinapses Imunológicas/imunologia , Sinapses Imunológicas/metabolismo , Masculino , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas , Ácidos Sulfônicos/uso terapêutico
5.
J Biol Chem ; 294(43): 15836-15849, 2019 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-31495784

RESUMO

Cholesterol esters are a key ingredient of foamy cells in atherosclerotic lesions; their formation is catalyzed by two enzymes: acyl-CoA:cholesterol acyltransferases (ACATs; also called sterol O-acyltransferases, or SOATs) ACAT1 and ACAT2. ACAT1 is present in all body cells and is the major isoenzyme in macrophages. Whether blocking ACAT1 benefits atherosclerosis has been under debate for more than a decade. Previously, our laboratory developed a myeloid-specific Acat1 knockout (KO) mouse (Acat1-M/-M), devoid of ACAT1 only in macrophages, microglia, and neutrophils. In previous work using the ApoE KO (ApoE-/-) mouse model for early lesions, Acat1-M/-M significantly reduced lesion macrophage content and suppressed atherosclerosis progression. In advanced lesions, cholesterol crystals become a prominent feature. Here we evaluated the effects of Acat1-M/-M in the ApoE KO mouse model for more advanced lesions and found that mice lacking myeloid Acat1 had significantly reduced lesion cholesterol crystal contents. Acat1-M/-M also significantly reduced lesion size and macrophage content without increasing apoptotic cell death. Cell culture studies showed that inhibiting ACAT1 in macrophages caused cells to produce less proinflammatory responses upon cholesterol loading by acetyl low-density lipoprotein. In advanced lesions, Acat1-M/-M reduced but did not eliminate foamy cells. In advanced plaques isolated from ApoE-/- mice, immunostainings showed that both ACAT1 and ACAT2 are present. In cell culture, both enzymes are present in macrophages and smooth muscle cells and contribute to cholesterol ester biosynthesis. Overall, our results support the notion that targeting ACAT1 or targeting both ACAT1 and ACAT2 in macrophages is a novel strategy to treat advanced lesions.


Assuntos
Aterosclerose/enzimologia , Aterosclerose/prevenção & controle , Inflamação/patologia , Macrófagos Peritoneais/enzimologia , Células Mieloides/enzimologia , Esterol O-Aciltransferase/metabolismo , Animais , Apolipoproteínas E , Apoptose , Aterosclerose/patologia , Colesterol/metabolismo , Ciclo-Oxigenase 2/metabolismo , Modelos Animais de Doenças , Deleção de Genes , Inativação Gênica , Hidroxicolesteróis/metabolismo , Lipoproteínas LDL/metabolismo , Camundongos , Camundongos Knockout , Músculo Liso Vascular/patologia , Células Mieloides/patologia , Miócitos de Músculo Liso/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Células RAW 264.7
6.
J Cell Sci ; 129(16): 3104-14, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27358480

RESUMO

Excitatory amino acid transporter type 3 (EAAT3, also known as SLC1A1) is a high-affinity, Na(+)-dependent glutamate carrier that localizes primarily within the cell and at the apical plasma membrane. Although previous studies have reported proteins and sequence regions involved in EAAT3 trafficking, the detailed molecular mechanism by which EAAT3 is distributed to the correct location still remains elusive. Here, we identify that the YVNGGF sequence in the C-terminus of EAAT3 is responsible for its intracellular localization and apical sorting in rat hepatoma cells CRL1601 and Madin-Darby canine kidney (MDCK) cells, respectively. We further demonstrate that Numb, a clathrin adaptor protein, directly binds the YVNGGF motif and regulates the localization of EAAT3. Mutation of Y503, N505 and F508 within the YVNGGF motif to alanine residues or silencing Numb by use of small interfering RNA (siRNA) results in the aberrant localization of EAAT3. Moreover, both Numb and the YVNGGF motif mediate EAAT3 endocytosis in CRL1601 cells. In summary, our study suggests that Numb is a pivotal adaptor protein that mediates the subcellular localization of EAAT3 through binding the YxNxxF (where x stands for any amino acid) motif.


Assuntos
Transportador 3 de Aminoácido Excitatório/química , Transportador 3 de Aminoácido Excitatório/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Motivos de Aminoácidos , Animais , Cães , Endocitose , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Masculino , Camundongos Endogâmicos BALB C , Mutação/genética , Ligação Proteica , Transporte Proteico , Ratos , Relação Estrutura-Atividade , Frações Subcelulares/metabolismo
7.
Hepatology ; 65(6): 1936-1947, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28027595

RESUMO

Obesity is a critical risk factor for hepatocellular carcinoma (HCC). However, it remains unknown whether inhibition of de novo lipid biosynthesis can suppress HCC. In this study, we blocked the sterol regulatory element-binding protein (SREBP) pathway, one of the key determinants of lipid homeostasis, by ablating 78-kDa cell-surface glycoprotein or SREBP cleavage-activating protein in hepatocytes, as well as by administering a chemical compound called betulin. We found that either genetically or pharmacologically inhibiting the SREBP pathway dramatically reduced diethylnitrosamine-induced HCC progression by down-regulating tumor-promoting cytokines, including interleukin (IL)-6, tumor necrosis factor alpha, and IL-1ß. CONCLUSION: Inhibition of de novo lipid biosynthesis by suppressing the SREBP pathway prevents HCC. This study identifies a previously underappreciated role of the SREBP pathway in HCC and suggests a novel metabolic strategy to control liver cancer. (Hepatology 2017;65:1936-1947).


Assuntos
Carcinogênese/patologia , Carcinoma Hepatocelular/patologia , Interleucina-1beta/metabolismo , Neoplasias Hepáticas/patologia , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Fator de Necrose Tumoral alfa/metabolismo , Animais , Carcinoma Hepatocelular/fisiopatologia , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica , Hepatócitos/metabolismo , Hepatócitos/patologia , Inflamação/patologia , Inflamação/prevenção & controle , Neoplasias Hepáticas/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias Experimentais , Obesidade/complicações , Obesidade/patologia , Ligação Proteica/genética , Distribuição Aleatória , Reação em Cadeia da Polimerase em Tempo Real/métodos , Valores de Referência , Fatores de Risco , Células Tumorais Cultivadas
8.
J Lipid Res ; 58(3): 512-518, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28053186

RESUMO

Niemann-Pick type C (NPC) disease is a fatal inherited neurodegenerative disorder caused by loss-of-function mutations in the NPC1 or NPC2 gene. There is no effective way to treat NPC disease. In this study, we used adeno-associated virus (AAV) serotype 9 (AAV9) to deliver a functional NPC1 gene systemically into NPC1-/- mice at postnatal day 4. One single AAV9-NPC1 injection resulted in robust NPC1 expression in various tissues, including brain, heart, and lung. Strikingly, AAV9-mediated NPC1 delivery significantly promoted Purkinje cell survival, restored locomotor activity and coordination, and increased the lifespan of NPC1-/- mice. Our work suggests that AAV-based gene therapy is a promising means to treat NPC disease.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética , Doença de Niemann-Pick Tipo C/genética , Doença de Niemann-Pick Tipo C/terapia , Proteínas/genética , Animais , Encéfalo/metabolismo , Sobrevivência Celular/genética , Dependovirus/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Locomoção/genética , Pulmão/metabolismo , Camundongos , Miocárdio/metabolismo , Proteína C1 de Niemann-Pick , Doença de Niemann-Pick Tipo C/patologia , Proteínas/administração & dosagem , Células de Purkinje/metabolismo , Células de Purkinje/patologia , Proteínas de Transporte Vesicular/administração & dosagem , Proteínas de Transporte Vesicular/genética
9.
Biochem Biophys Res Commun ; 479(4): 628-635, 2016 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-27697530

RESUMO

BACKGROUND: Plasma levels of low-density lipoprotein cholesterol (LDL-C) are a major risk factor for cardiovascular disease and are influenced by both heredity and dietary habits. The Niemann-Pick C1 like 1 (NPC1L1) protein mediates efficient dietary cholesterol absorption and contributes to variations in human LDL-C levels. METHODS: In the present study, using high throughput sequencing we identified three non-synonymous (NS) variations and 64 synonymous variations in the NPC1L1 gene from subsets of Chinese Han, Uygur and Kazakh populations with high or low LDL-C. Subsequently, three NS variations encoding R174H, V177I and V1284L substitutions were observed only in Uygur and Kazakh individuals with limited maximal plasma LDL-C levels. RESULTS: In further experiments, we investigated cholesterol-regulated recycling and glycosylation and stability of these NS NPC1L1 variants. However, no significant differences between WT and variant NPC1L1 proteins were observed using in vivo assays in mouse livers with adenovirus-mediated expression, demonstrating that none of the three NPC1L1 NS variants caused decreased uptake of biliary cholesterol. CONCLUSIONS: Simultaneously, these data indicate that R174H, V177I and V1284L NPC1L1 variations in high or low LDL-C individuals may not directly influence cholesterol absorption by NPC1L1.


Assuntos
VLDL-Colesterol/sangue , Etnicidade/genética , Variação Genética , Hipercolesterolemia/genética , Proteínas de Membrana/genética , Adulto , Animais , Linhagem Celular Tumoral , China/etnologia , VLDL-Colesterol/genética , VLDL-Colesterol/metabolismo , Feminino , Humanos , Hipercolesterolemia/sangue , Reabsorção Intestinal/genética , Cazaquistão/etnologia , Fígado/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras , Camundongos Endogâmicos ICR , Pessoa de Meia-Idade , Fases de Leitura Aberta/genética , Ratos
10.
Org Biomol Chem ; 14(2): 747-751, 2016 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-26584338

RESUMO

A series of pyripyropene A-based compounds were designed and synthesized by opening the upper section of the A-ring, which significantly simplifies the structure and synthesis from commercially available starting materials. Representative compound (-)-3 exhibited potent activity against ACAT2 and greater selectivity for ACAT2 than for ACAT1.


Assuntos
Desenho de Fármacos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Piridinas/farmacologia , Sesquiterpenos/farmacologia , Esterol O-Aciltransferase/antagonistas & inibidores , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/química , Humanos , Estrutura Molecular , Piridinas/síntese química , Piridinas/química , Sesquiterpenos/síntese química , Sesquiterpenos/química , Esterol O-Aciltransferase/metabolismo , Relação Estrutura-Atividade , Esterol O-Aciltransferase 2
12.
J Biol Chem ; 289(48): 33689-700, 2014 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-25331956

RESUMO

The uptake of circulating low density lipoproteins (LDL) is mediated by LDL receptor (LDLR) through clathrin-dependent endocytosis. At the early stage of this process, adaptor proteins ARH and Dab2 specifically bind the endocytic signal motif in LDLR and recruit clathrin/AP2 to initiate internalization. On the other hand, intestinal cholesterol is absorbed by Niemann-Pick C1-Like 1 (NPC1L1) through clathrin-dependent endocytosis. Another adaptor protein, Numb recognizes the endocytic motif in NPC1L1 C terminus and couples NPC1L1 to endocytic machinery. The ARH, Dab2, and Numb proteins contain a homogeneous phosphotyrosine binding (PTB) domain that directly binds endocytic motifs. Because ARH, Dab2, and Numb are all PTB domain family members, the emerging mystery is whether these adaptors act complementally in LDLR and NPC1L1 endocytosis. Here, we found that ARH and Dab2 did not bind NPC1L1 and were not required for NPC1L1 internalization. Similarly, Numb lacked the ability to interact with the LDLR C terminus and was dispensable for LDL uptake. Only the Numb isoforms with shorter PTB domain could facilitate NPC1L1 endocytosis. Besides the reported function in intestinal cholesterol absorption, Numb also mediated cholesterol reabsorption from bile in liver. We further identified a Numb variant with G595D substitution in humans of low blood LDL-cholesterol. The G595D substitution impaired NPC1L1 internalization and cholesterol reabsorption, due to attenuating affinity of Numb to clathrin/AP2. These results demonstrate that Numb specifically regulates NPC1L1-mediated cholesterol absorption both in human intestine and liver, distinct from ARH and Dab2, which selectively participate in LDLR-mediated LDL uptake.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , LDL-Colesterol/metabolismo , Endocitose/fisiologia , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptores de LDL/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Substituição de Aminoácidos , Animais , Proteínas Reguladoras de Apoptose , Transporte Biológico Ativo/fisiologia , Linhagem Celular Tumoral , LDL-Colesterol/genética , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana Transportadoras , Mutação de Sentido Incorreto , Proteínas do Tecido Nervoso/genética , Ratos , Receptores de LDL/genética , Proteínas Supressoras de Tumor/genética
13.
Proc Natl Acad Sci U S A ; 108(2): 551-6, 2011 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-21187433

RESUMO

Dietary absorption is a major way for mammals to obtain cholesterol, which is mediated by Niemann-Pick C1-like 1 (NPC1L1) via vesicular endocytosis. One fundamental question in this process is how free cholesterol is efficiently taken up through the internalization of NPC1L1. Using exogenously expressed NPC1L1-EGFP, we show that the lipid raft proteins flotillins associate with NPC1L1 and their localization is regulated by NPC1L1 during intracellular trafficking. Furthermore, flotillins are essential for NPC1L1-mediated cellular cholesterol uptake, biliary cholesterol reabsorption, and the regulation of lipid levels in mice. Together with NPC1L1, they form cholesterol-enriched membrane microdomains, which function as carriers for bulk of cholesterol. The hypocholesterolemic drug ezetimibe disrupts the association between NPC1L1 and flotillins, which blocks the formation of the cholesterol-enriched microdomains. Our findings reveal a functional role of flotillins in NPC1L1-mediated cholesterol uptake and elucidate the formation of NPC1L1-flotillins-postive cholesterol-enriched membrane microdomains as a mechanism for efficient cholesterol absorption.


Assuntos
Colesterol/metabolismo , Proteínas de Membrana/metabolismo , Doenças de Niemann-Pick/metabolismo , Animais , Anticolesterolemiantes/farmacologia , Azetidinas/farmacologia , Colesterol/química , Ezetimiba , Proteínas de Fluorescência Verde/metabolismo , Homeostase , Humanos , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Microscopia Confocal/métodos , Microscopia de Fluorescência/métodos , Ratos
14.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 45(2): 278-80, 2014 Mar.
Artigo em Zh | MEDLINE | ID: mdl-24749357

RESUMO

OBJECTIVE: To detect changes of thyroid stimulating hormone (TSH) with gender, age and levels of thyroid peroxidase antibodies (TPO-Ab) in patients with subclinical hypothyroidism, and to establish composite reference intervals for TSH and free thyroxine (FT4) in determination of subclinical hypothyroidism. METHODS: From Oct. 2011 to July 2012, 7 964 healthy people (males: 4 789, females: 3 175) undergoing medical examinations were recruited. Their serum levels of TSH and FT4 were determined. Of those participants, 794 were also tested for TPO-Ab. The serum TSH and FT4 data were transformed into normal distributions, with outliers being eliminated and a correction for skewness (0. 909 and 0. 384, respectively) and kurtosis (2.605 and 1.947, respectively). The composite reference intervals were established according to the Mahalonobis distance formula. RESULTS: Serum TSH increased with age and TPO-Ab. Using the conventional reference standards, 358 participants were identified with subclinical hypothyroidism, which included 230 at 41-70 years of age and 43 showing TPO-Ab positive. In contrast, using composite reference intervals, 301 participants were identified with subclinical hypothyroidism, which included 142 at 41-70 years of age and 25 showing TPO-Ab. CONCLUSION: The conventional cutoff values for FT4 and TSH separately lead to overestimation of the prevalence of subclinical thyroid disease.


Assuntos
Hipotireoidismo/diagnóstico , Tireotropina/sangue , Tiroxina/sangue , Adulto , Idoso , Autoanticorpos/sangue , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Valores de Referência , Testes de Função Tireóidea
15.
Phytomedicine ; 132: 155835, 2024 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-38968791

RESUMO

BACKGROUND: Iron deposition and ferroptosis are involved in ischemic stroke injury, but the choice of drugs for treatment is limited. PURPOSE: To investigate the potential neuroprotective effects of Rosmarinic acid (RosA) encapsulated within nanoliposomes (RosA-LIP) on ischemic stroke. METHODS: Wild-type (WT) and TfR1EC cKO (specific knockout of the TfR1 gene in BMECs) mice used to establish a dMCAO model, with simultaneous administration of RosA-LIP (20 mg/kg/d, i.p.) or RosA (20 mg/kg/d, i.p.). RESULTS: The successful synthesis of RosA-LIP resulted in enhanced stability and precise delivery in both the serum and brain. The administration of RosA-LIP effectively mitigated ischemia-induced behavioral abnormalities and pathological damage. RosA-LIP inhibited ferroptosis by ameliorating mitochondrial abnormalities, increasing GPX4 levels, and decreasing ACSL4/LPCAT3/Lox-dependent lipid peroxidation. RosA-LIP effectively improved blood‒brain barrier (BBB) permeability, increased tight junctions (TJs) protein expression and reduced iron levels in ischemic tissue and brain microvascular endothelial cells (BMECs) by modulating FPN1 and TfR1 levels. Furthermore, RosA-LIP suppressed TfR1 to attenuate ACSL4/LPCAT3/Lox-mediated ferroptosis in TfR1EC cKO mice subjected to dMCAO. CONCLUSION: RosA-LIP effectively increased the brain level of RosA and protected against ferroptosis through the regulation of TfR1 in BMECs.

16.
Artigo em Inglês | MEDLINE | ID: mdl-38175414

RESUMO

The objective of this study is to examine the potential protective effect of rosmarinic acid (RosA) encapsulated within nanoliposomes (RosA-LIP) on hepatic damage induced by iron overload. The characteristics, stability, and release of RosA-LIP in vitro were identified. The mice were randomly assigned to five groups: Control, Model, Model+DFO (DFO), Model+RosA (RosA), and Model+RosA-LIP (RosA-LIP). The iron overload model was induced by administering iron dextran (i.p.). The DFO, RosA, and RosA-LIP groups received iron dextran and were subsequently treated with DFO, RosA, and RosA-LIP for 14 days. We developed a novel formulation of RosA-LIP that exhibited stability and controlled release properties. Firstly, RosA-LIP improved liver function and ameliorated pathological changes in a mouse model of iron overload. Secondly, RosA-LIP demonstrated the ability to enhance the activities of T-SOD, GSH-Px, and CAT, while reducing the levels of MDA and 4-HNE, thereby effectively mitigating oxidative stress damage induced by iron overload. Thirdly, RosA-LIP reduced hepatic iron levels by downregulating FTL, FTH, and TfR1 levels. Additionally, RosA-LIP exerted a suppressive effect on hepcidin expression through the BMP6-SMAD1/5/8 signaling pathway. Furthermore, RosA-LIP upregulated FPN1 expression in both the liver and duodenum, thereby alleviating iron accumulation in these organs in mice with iron overload. Notably, RosA exhibited a comparable iron chelation effect, and RosA-LIP demonstrated superior efficacy in mitigating liver damage induced by excessive iron overload. RosA-LIP exhibited favorable sustained release properties, targeted delivery, and efficient protection against iron overload-induced liver damage. A schematic representation of the proposed protective mechanism of rosmarinic acid liposome during iron overload. Once RosA-LIP is transported into cells, RosA is released. On the one hand, RosA attenuates the BMP6-SMAD1/5/8-SMAD4 signaling pathway activation, leading to inhibiting hepcidin transcription. Then, the declined hepcidin contacted the inhibitory effect of FPN1 in hepatocytes and duodenum, increasing iron mobilization. On the other hand, RosA inhibits TfR1 and ferritin expression, which decreases excessive iron and oxidative damage.

17.
Cell Metab ; 6(2): 115-28, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17681147

RESUMO

The membrane-anchored ubiquitin ligase gp78 promotes degradation of misfolded endoplasmic reticulum (ER) proteins and sterol-regulated degradation of HMG-CoA reductase. It was known previously that Ufd1 plays a critical role in ER-associated degradation (ERAD) together with Npl4 and VCP. The VCP-Ufd1-Npl4 complex recognizes polyubiquitin chains and transfers the ubiquitinated proteins to the proteasome. Here we show that Ufd1 directly interacts with gp78 and functions as a cofactor. Ufd1 enhances the E3 activity of gp78, accelerates the ubiquitination and degradation of reductase, and eventually promotes receptor-mediated uptake of low-density lipoprotein. Furthermore, we demonstrate that the monoubiquitin-binding site in Ufd1 is required for the enhancement of gp78 activity and that the polyubiquitin-binding site in Ufd1 is critical for a postubiquitination step in ERAD. In summary, our study identifies Ufd1 as a cofactor of gp78, reveals an unappreciated function of Ufd1 in the ubiquitination reaction during ERAD, and illustrates that Ufd1 plays a critical role in cholesterol metabolism.


Assuntos
Colesterol/metabolismo , Hidroximetilglutaril-CoA Redutases/metabolismo , Proteínas/metabolismo , Receptores de Citocinas/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Adaptadoras de Transporte Vesicular , Aminoácidos , Animais , Sítios de Ligação , Células CHO , Linhagem Celular , Cricetinae , Cricetulus , Estabilidade Enzimática , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Lipoproteínas LDL/metabolismo , Modelos Biológicos , Ligação Proteica , Mapeamento de Interação de Proteínas , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Receptores do Fator Autócrino de Motilidade , Receptores de Citocinas/química , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/química
18.
J Biol Chem ; 286(41): 35933-35942, 2011 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-21844200

RESUMO

Niemann-Pick C1-like 1 (NPC1L1) is a multi-transmembrane protein that mediates the absorption of dietary and biliary cholesterol through vesicular endocytosis. The subcellular localization of NPC1L1 is regulated by cholesterol. Cholesterol depletion induces the transport of NPC1L1 to plasma membrane (PM) from endocytic recycling compartment that requires MyoVb·Rab11a·Rab11-FIP2 triple complex, and cholesterol-replenishment renders the internalization of NPC1L1 together with cholesterol. Here, we find that GTP-bound Cdc42 interacts with NPC1L1. Cholesterol depletion regulates the activation of Cdc42 and enhances NPC1L1-Cdc42 interaction. Overexpression of constitutive GTP-bound Cdc42 mutant form or knockdown of Cdc42 inhibits the transport of NPC1L1 to the PM and disturbs the cholesterol-regulated binding of NPC1L1 to Rab11a, MyoVb, and actin. Knockdown of Cdc42 downstream effectors N-WASP or Arp3 also leads to the similar results. In liver-specific Cdc42 knock-out (Cdc42 LKO) mice, NPC1L1 fails to localize to bile canaliculi, and the biliary cholesterol cannot be efficiently reabsorbed. These results indicate that Cdc42 controls the cholesterol-regulated transport and localization of NPC1L1, and plays a role in cholesterol absorption.


Assuntos
Membrana Celular/metabolismo , Colesterol/metabolismo , Endocitose/fisiologia , Proteínas de Membrana Transportadoras/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteína 3 Relacionada a Actina/genética , Proteína 3 Relacionada a Actina/metabolismo , Actinas/genética , Actinas/metabolismo , Animais , Canalículos Biliares/metabolismo , Transporte Biológico Ativo/fisiologia , Linhagem Celular Tumoral , Membrana Celular/genética , Colesterol/genética , Ativação Enzimática/fisiologia , Proteínas de Membrana Transportadoras/genética , Camundongos , Camundongos Knockout , Ratos , Proteína Neuronal da Síndrome de Wiskott-Aldrich/genética , Proteína Neuronal da Síndrome de Wiskott-Aldrich/metabolismo , Proteína cdc42 de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo
19.
J Biol Chem ; 286(9): 7397-408, 2011 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-21189420

RESUMO

Niemann-Pick C1-like 1 (NPC1L1) is an essential protein for dietary cholesterol absorption. Nonsynonymous (NS) variants of NPC1L1 in humans have been suggested to associate with cholesterol absorption variations. However, information concerning the characteristics and mechanism of these variants in cholesterol uptake is limited. In this study, we analyzed the cholesterol uptake ability of the 19 reported NS variants of NPC1L1 identified from cholesterol low absorbers. Among these variants, L110F, R306C, A395V, G402S, T413M, R693C, R1214H, and R1268H could partially mediate cellular cholesterol uptake and were categorized as partially dysfunctional variants. The other 11 variants including T61M, N132S, D398G, R417W, G434R, T499M, S620C, I647N, G672R, S881L, and R1108W could barely facilitate cholesterol uptake, and were classified into the severely dysfunctional group. The partially dysfunctional variants showed mild defects in one or multiple aspects of cholesterol-regulated recycling, subcellular localization, glycosylation, and protein stability. The severely dysfunctional ones displayed remarkable defects in all these aspects and were rapidly degraded through the ER-associated degradation (ERAD) pathway. In vivo analyses using adenovirus-mediated expression in mouse liver confirmed that the S881L variant failed to localize to liver canalicular membrane, and the mice showed defects in biliary cholesterol re-absorption, while the G402S variant appeared to be similar to wild-type NPC1L1 in mouse liver. This study suggests that the dysfunction of the 19 variants on cholesterol absorption is due to the impairment of recycling, subcellular localization, glycosylation, or stability of NPC1L1.


Assuntos
Colesterol na Dieta/farmacocinética , Variação Genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Animais , Bile/metabolismo , Endocitose/fisiologia , Retículo Endoplasmático/metabolismo , Glicosilação , Humanos , Fígado/metabolismo , Proteínas de Membrana Transportadoras , Camundongos , Camundongos Endogâmicos ICR , Polimorfismo Genético , Estabilidade Proteica , Transporte Proteico/fisiologia
20.
J Biol Chem ; 286(28): 25088-97, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21602275

RESUMO

Niemann-Pick C1-like 1 (NPC1L1) is a multitransmembrane protein playing a crucial role in dietary and biliary cholesterol absorption. Cholesterol promotes the formation and endocytosis of NPC1L1-flotillin-cholesterol membrane microdomains, which is an early step in cholesterol uptake. How cholesterol is sensed in this step is unknown. Here, we find that the N-terminal domain (NTD) of NPC1L1 binds cholesterol. Mutation of residue Leu-216 in NPC1L1-NTD eliminates cholesterol binding, decreases the formation of NPC1L1-flotillin-cholesterol membrane microdomains, and prevents NPC1L1-mediated cholesterol uptake in culture cells and mice livers. NPC1L1-NTD specifically binds cholesterol but not plant sterols, which may account for the selective cholesterol absorption in intestine. Furthermore, 25- or 27-hydroxycholesterol competes with cholesterol to bind NPC1L1-NTD and inhibits the cholesterol induced endocytosis of NPC1L1. Together, these results demonstrate that plasma membrane-localized NPC1L1 binds exogenous cholesterol via its NTD, and facilitates the formation of NPC1L1-flotillin-cholesterol membrane microdomains that are then internalized into cells through the clathrin-AP2 pathway. Our study uncovers the mechanism of cholesterol sensing by NPC1L1 and proposes a mechanism for selective cholesterol absorption.


Assuntos
Colesterol/metabolismo , Endocitose/fisiologia , Fígado/metabolismo , Microdomínios da Membrana/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Absorção/fisiologia , Complexo 2 de Proteínas Adaptadoras/genética , Complexo 2 de Proteínas Adaptadoras/metabolismo , Animais , Transporte Biológico Ativo/fisiologia , Colesterol/genética , Clatrina/genética , Clatrina/metabolismo , Células HEK293 , Humanos , Microdomínios da Membrana/genética , Proteínas de Membrana/genética , Proteínas de Membrana Transportadoras/genética , Camundongos , Ligação Proteica , Estrutura Terciária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA