Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Mol Cell Proteomics ; 21(1): 100174, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34763061

RESUMO

The lysosome represents a central degradative compartment of eukaryote cells, yet little is known about the biogenesis and function of this organelle in parasitic protists. Whereas the mannose 6-phosphate (M6P)-dependent system is dominant for lysosomal targeting in metazoans, oligosaccharide-independent sorting has been reported in other eukaryotes. In this study, we investigated the phagolysosomal proteome of the human parasite Trichomonas vaginalis, its protein targeting and the involvement of lysosomes in hydrolase secretion. The organelles were purified using Percoll and OptiPrep gradient centrifugation and a novel purification protocol based on the phagocytosis of lactoferrin-covered magnetic nanoparticles. The analysis resulted in a lysosomal proteome of 462 proteins, which were sorted into 21 classes. Hydrolases represented the largest functional class and included proteases, lipases, phosphatases, and glycosidases. Identification of a large set of proteins involved in vesicular trafficking (80) and turnover of actin cytoskeleton rearrangement (29) indicate a dynamic phagolysosomal compartment. Several cysteine proteases such as TvCP2 were previously shown to be secreted. Our experiments showed that secretion of TvCP2 was strongly inhibited by chloroquine, which increases intralysosomal pH, thus indicating that TvCP2 secretion occurs through lysosomes rather than the classical secretory pathway. Unexpectedly, we identified divergent homologues of the M6P receptor TvMPR in the phagolysosomal proteome, although T. vaginalis lacks enzymes for M6P formation. To test whether oligosaccharides are involved in lysosomal targeting, we selected the lysosome-resident cysteine protease CLCP, which possesses two glycosylation sites. Mutation of any of the sites redirected CLCP to the secretory pathway. Similarly, the introduction of glycosylation sites to secreted ß-amylase redirected this protein to lysosomes. Thus, unlike other parasitic protists, T. vaginalis seems to utilize glycosylation as a recognition marker for lysosomal hydrolases. Our findings provide the first insight into the complexity of T. vaginalis phagolysosomes, their biogenesis, and role in the unconventional secretion of cysteine peptidases.


Assuntos
Cisteína Proteases , Trichomonas vaginalis , Cisteína/metabolismo , Cisteína Proteases/metabolismo , Humanos , Lisossomos/metabolismo , Peptídeo Hidrolases/metabolismo , Fagossomos/metabolismo , Proteômica , Trichomonas vaginalis/metabolismo
2.
Sci Rep ; 11(1): 19634, 2021 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-34608194

RESUMO

The persistent increase of multidrug-resistant (MDR) Mycobacterium tuberculosis (Mtb) infections negatively impacts Tuberculosis treatment outcomes. Host-directed therapies (HDT) pose an complementing strategy, particularly since Mtb is highly successful in evading host-defense by manipulating host-signaling pathways. Here, we screened a library containing autophagy-modulating compounds for their ability to inhibit intracellular Mtb-bacteria. Several active compounds were identified, including two drugs of the diphenylbutylpiperidine-class, Fluspirilene and Pimozide, commonly used as antipsychotics. Both molecules inhibited intracellular Mtb in pro- as well as anti-inflammatory primary human macrophages in a host-directed manner and synergized with conventional anti-bacterials. Importantly, these inhibitory effects extended to MDR-Mtb strains and the unrelated intracellular pathogen, Salmonella enterica serovar Typhimurium (Stm). Mechanistically Fluspirilene and Pimozide were shown to regulate autophagy and alter the lysosomal response, partly correlating with increased bacterial localization to autophago(lyso)somes. Pimozide's and Fluspirilene's efficacy was inhibited by antioxidants, suggesting involvement of the oxidative-stress response in Mtb growth control. Furthermore, Fluspirilene and especially Pimozide counteracted Mtb-induced STAT5 phosphorylation, thereby reducing Mtb phagosome-localized CISH that promotes phagosomal acidification. In conclusion, two approved antipsychotic drugs, Pimozide and Fluspirilene, constitute highly promising and rapidly translatable candidates for HDT against Mtb and Stm and act by modulating the autophagic/lysosomal response by multiple mechanisms.


Assuntos
Antibacterianos/farmacologia , Antipsicóticos/farmacologia , Antituberculosos/farmacologia , Reposicionamento de Medicamentos , Mycobacterium tuberculosis/efeitos dos fármacos , Salmonella enterica/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Linhagem Celular , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Ensaios de Triagem em Larga Escala , Humanos , Lisossomos/metabolismo , Testes de Sensibilidade Microbiana , Modelos Biológicos , Fagossomos/metabolismo , Pimozida/farmacologia , Infecções por Salmonella/tratamento farmacológico , Infecções por Salmonella/microbiologia , Bibliotecas de Moléculas Pequenas , Tuberculose/tratamento farmacológico , Tuberculose/microbiologia
3.
Anal Chem ; 92(23): 15437-15444, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33201688

RESUMO

Proximity-based in situ labeling techniques offer a unique way to capture both stable and transient protein-protein and protein-organelle interactions. Combining this technology with mass spectrometry (MS)-based proteomics allows us to obtain snapshots of molecular microenvironments with nanometer resolution, facilitating the discovery of complex and dynamic protein networks. However, a number of technical challenges still exist, such as interferences from endogenously biotinylated proteins and other highly abundant bystanders, how to select the proper controls to minimize false discoveries, and experimental variations among biological/technical replicates. Here, we developed a new method to capture the proteomic microenvironment of the neuronal endolysosomal network by knocking in (KI) an engineered ascorbate peroxidase (APEX) gene to the endogenous locus of lysosome-associated membrane protein 1 (LAMP1). We found that normalizing proximity labeling proteomics data to the endogenously biotinylated protein (PCCA) can greatly reduce variations and enable fair comparisons among different batches of APEX labeling and different APEX probes. We conducted a comparative evaluation between this KI-LAMP1-APEX method and our two overexpression LAMP1-APEX probes, achieving complementary coverage of both known and new lysosomal membrane and lysosomal-interacting proteins in human iPSC-derived neurons. To summarize, this study demonstrated new analytical tools to characterize lysosomal functions and microenvironment in human neurons and filled critical gaps in the field for designing and optimizing proximity labeling proteomic experiments.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Neurônios/citologia , Fagossomos/metabolismo , Proteômica/métodos , Ascorbato Peroxidases/genética , Técnicas de Introdução de Genes , Humanos , Proteína 1 de Membrana Associada ao Lisossomo/metabolismo , Neurônios/metabolismo , Coloração e Rotulagem
4.
Cell Calcium ; 91: 102269, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32818767

RESUMO

Lysosomes are generally thought to be required only for the late stages of phagosome maturation, providing the proton pumps (V-ATPases) and hydrolases needed to acidify and degrade the ingested prey. A recent paper by Davis et al. (EMBO J. [2020], doi:10.15252/embj.2019104058) reports the involvement of lysosomes at a much earlier stage, namely in scission of phagosomes from the plasma membrane. Here we analyze these findings, highlighting a number of unexpected observations and unresolved questions.


Assuntos
Lisossomos/metabolismo , Fagocitose , Animais , Calcineurina/metabolismo , Cálcio/metabolismo , Humanos , NADP/análogos & derivados , NADP/metabolismo , Fagossomos/metabolismo
5.
J Exp Clin Cancer Res ; 38(1): 353, 2019 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-31412953

RESUMO

BACKGROUND: Tubeimoside-I (TBM), a plant-derived bioactive compound, shows antitumor activity in different tumors and can enhance the efficacy of chemotherapeutic agents. However, the detail mechanism underlying remains to be elucidated. METHODS: The cytotoxic potential of TBM towards CRC cells was examined by CCK8 assay, colony formation, LDH release assay, flow cytometry method and Western blots. The ROS levels, autophagy, apoptosis, chemosensitivity to 5-FU or DOX, etc. were determined between control and TBM-treated CRC cells. RESULTS: In this study, we found that TBM could inhibit proliferation and induce apoptosis in colorectal cancer (CRC) cells. Intriguingly, TBM treatment could either promote autophagy initiation by ROS-induced AMPK activation, or block autophagy flux through inhibiting lysosomal hydrolytic enzymes, which leaded to massive impaired autophagylysosomes accumulation. Administration of autophagy initiation inhibitor (3-MA or selective ablation of autophagy related proteins) relieves TBM-induced CRC suppression, while combination use of autophagy flux inhibitor chloroquine (CQ) slightly augments TBM-induced cell death, suggesting that impaired autophagylysosomes accumulation contributes to TBM-induced growth inhibition in CRC cells. Notably, as an autophagy flux inhibitor, TBM works synergistically with 5-fluorouracil (5-FU) or doxorubicin (DOX) in CRC suppression. CONCLUSION: Together, our study provides new insights regarding the anti-tumor activity of TBM against CRC, and established potential applications of TBM for CRC combination therapies in clinic.


Assuntos
Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Medicamentos de Ervas Chinesas/farmacologia , Fagossomos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Saponinas/farmacologia , Triterpenos/farmacologia , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sinergismo Farmacológico , Fluoruracila/farmacologia , Humanos , Lisossomos/metabolismo , Proteólise/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
6.
PLoS One ; 14(3): e0213150, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30830942

RESUMO

In nature, many plants or their extracted compounds have been found to possess anti-inflammatory features and therapeutic properties against infectious as well as non-infectious diseases, including cancer. In this study, we analysed the immunomodulatory effects on innate immune cells of hydroalcoholic extract from Origanum vulgare L. ssp. hirtum (HyE-Ov), a plant traditionally known for its anti-oxidative properties. The effects of HyE-Ov were tested on human monocyte derived dendritic cells (DC), type-1 (M1) and type-2 macrophages (M2) infected with M. bovis Bacille Calmette-Guérin (BCG), used as a model of persistent intracellular bacterium. DC, M1 and M2 treated with HyE-Ov significantly enhanced their mycobactericidal activity, which was associated with phagosomal acidification in M1 and M2 and increase of phagosomal, but not mitochondrial ROS production in M1, M2, and DC. Treatment of BCG-infected DC with HyE-Ov significantly reduced TNF-α and IL-12 production and increased TGF-ß synthesis. Finally, experiments were repeated using eight different HPLC fractions of HyE-Ov. Results showed that the capability to activate anti-microbial and anti-inflammatory response is shared by different fractions, suggesting that diverse bioactive molecules are present within the hydroalcoholic extract. Altogether, these results show that HyE-Ov promotes anti-mycobacterial innate immunity and limits inflammatory response in vitro and suggest that this plant extract may be exploitable as phytocomplex or nutraceutical for novel host-directed therapeutic approaches.


Assuntos
Álcoois/farmacologia , Anti-Infecciosos/farmacologia , Anti-Inflamatórios/farmacologia , Células Dendríticas/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Mycobacterium bovis/efeitos dos fármacos , Origanum/química , Álcoois/química , Anti-Infecciosos/química , Anti-Inflamatórios/química , Células Cultivadas , Células Dendríticas/citologia , Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Voluntários Saudáveis , Humanos , Imunidade Inata/efeitos dos fármacos , Interleucina-2/metabolismo , Macrófagos/citologia , Macrófagos/imunologia , Macrófagos/microbiologia , Mycobacterium bovis/patogenicidade , Fagossomos/metabolismo , Extratos Vegetais/química , Extratos Vegetais/farmacologia , Fator de Crescimento Transformador beta/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
7.
Nat Commun ; 9(1): 3333, 2018 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-30127354

RESUMO

Mucormycosis is a life-threatening respiratory fungal infection predominantly caused by Rhizopus species. Mucormycosis has incompletely understood pathogenesis, particularly how abnormalities in iron metabolism compromise immune responses. Here we show how, as opposed to other filamentous fungi, Rhizopus spp. establish intracellular persistence inside alveolar macrophages (AMs). Mechanistically, lack of intracellular swelling of Rhizopus conidia results in surface retention of melanin, which induces phagosome maturation arrest through inhibition of LC3-associated phagocytosis. Intracellular inhibition of Rhizopus is an important effector mechanism, as infection of immunocompetent mice with swollen conidia, which evade phagocytosis, results in acute lethality. Concordantly, AM depletion markedly increases susceptibility to mucormycosis. Host and pathogen transcriptomics, iron supplementation studies, and genetic manipulation of iron assimilation of fungal pathways demonstrate that iron restriction inside macrophages regulates immunity against Rhizopus. Our findings shed light on the pathogenetic mechanisms of mucormycosis and reveal the role of macrophage-mediated nutritional immunity against filamentous fungi.


Assuntos
Interações Hospedeiro-Patógeno , Ferro/metabolismo , Pulmão/microbiologia , Macrófagos Alveolares/metabolismo , Rhizopus/fisiologia , Animais , Parede Celular/metabolismo , Regulação da Expressão Gênica , Macrófagos Alveolares/ultraestrutura , Melaninas/metabolismo , Camundongos Endogâmicos C57BL , Viabilidade Microbiana , Modelos Biológicos , Mucormicose/genética , Mucormicose/microbiologia , Mucormicose/patologia , Fagossomos/metabolismo , Fagossomos/ultraestrutura , Rhizopus/crescimento & desenvolvimento , Esporos Fúngicos/fisiologia
8.
Int J Biochem Cell Biol ; 70: 13-22, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26555754

RESUMO

The present study was performed to examine the induction of apoptotic cell death and autophagy by blue LED irradiation, and the contribution of autophagy to apoptosis in B cell lymphoma A20 and RAMOS cells exposed to blue LED. Irradiation with blue LED reduced cell viability and induced apoptotic cell death, as indicated by exposure of phosphatidylserine on the plasma outside membrane and fragmentation of DNA. Furthermore, the mitochondrial membrane potential increased, and apoptotic proteins (PARP, caspase 3, Bax, and bcl-2) were observed. In addition, the level of intracellular superoxide anion (O2(-)) gradually increased. Interestingly the formation of autophagosomes and level of LC3-II were increased in blue LED-irradiated A20 and RAMOS cells, but inhibited after pretreatment with 3-methyladenine (3-MA), widely used as an autophagy inhibitor. Inhibition of the autophagic process by pretreatment with 3-MA blocked blue LED irradiation-induced caspase-3 activation. Moreover, a significant reduction of both the early and late phases of apoptosis after transfection with ATG5 and beclin 1 siRNAs was shown by the annexin V/PI staining, indicating a crucial role of autophagy in blue LED-induced apoptosis in cells. Additionally, the survival rate of mice irradiated with blue LED after injection with A20 cells increased compared to the control group. Our data demonstrate that blue LED irradiation induces apoptosis via the mitochondrial-mediated pathway, in conjunction with autophagy. Further studies are needed to elucidate the precise mechanism of blue LED-induced immune cell death.


Assuntos
Autofagia/efeitos da radiação , Linfócitos B/efeitos da radiação , Linfoma de Células B/terapia , Mitocôndrias/efeitos da radiação , Fototerapia/métodos , Adenina/análogos & derivados , Adenina/farmacologia , Animais , Apoptose/efeitos da radiação , Linfócitos B/metabolismo , Linfócitos B/patologia , Caspase 3/genética , Caspase 3/metabolismo , Linhagem Celular Tumoral , Fragmentação do DNA/efeitos da radiação , Feminino , Humanos , Luz , Linfoma de Células B/genética , Linfoma de Células B/metabolismo , Linfoma de Células B/patologia , Potencial da Membrana Mitocondrial/efeitos da radiação , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Mitocôndrias/metabolismo , Transplante de Neoplasias , Fagossomos/metabolismo , Fagossomos/efeitos da radiação , Fototerapia/instrumentação , Poli(ADP-Ribose) Polimerases/genética , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Superóxidos/agonistas , Superóxidos/metabolismo , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
9.
Int J Mol Med ; 37(2): 309-18, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26647915

RESUMO

In a previous study by our group, we demonstrated that electroacupuncture (EA) activates the class I phosphoinositide 3-kinase (PI3K)/Akt signaling pathway. There is considerable evidence that the downstream mammalian target of rapamycin complex 1 (mTORC1) plays an important role in autophagy following ischemic stroke. The aim of the present study was to determine whether EA exerts a neuroprotective effect through mTORC1-mediated autophagy following ischemia/reperfusion injury. Our results revealed that EA at the LI11 and ST36 acupoints attenuated motor dysfunction, improved neurological deficit outcomes and decreased the infarct volumes. The number of autophagosomes, autolysosomes and lysosomes was decreased following treatment with EA. Simultaneously, the levels of the autophagosome membrane maker, microtubule-associated protein 1 light chain 3 beta (LC3B)Ⅱ/Ⅰ, Unc-51-like kinase 1 (ULK1), autophagy related gene 13 Atg13) and Beclin1 (ser14) were decreased, whereas mTORC1 expression was increased in the peri-infarct cortex. These results suggest that EA protects against ischemic stroke through the inhibition of autophagosome formation and autophagy, which is mediated through the mTORC1-ULK complex-Beclin1 pathway.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Eletroacupuntura/métodos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Traumatismo por Reperfusão/terapia , Acidente Vascular Cerebral/terapia , Pontos de Acupuntura , Animais , Proteínas Reguladoras de Apoptose/genética , Autofagia/genética , Proteína Homóloga à Proteína-1 Relacionada à Autofagia , Proteína Beclina-1 , Modelos Animais de Doenças , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lisossomos/genética , Lisossomos/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Fármacos Neuroprotetores , Fagossomos/genética , Fagossomos/metabolismo , Ratos , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia , Transdução de Sinais/genética , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/patologia , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
10.
Nature ; 527(7578): 323-8, 2015 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-26536114

RESUMO

Staphylococcus aureus is considered to be an extracellular pathogen. However, survival of S. aureus within host cells may provide a reservoir relatively protected from antibiotics, thus enabling long-term colonization of the host and explaining clinical failures and relapses after antibiotic therapy. Here we confirm that intracellular reservoirs of S. aureus in mice comprise a virulent subset of bacteria that can establish infection even in the presence of vancomycin, and we introduce a novel therapeutic that effectively kills intracellular S. aureus. This antibody-antibiotic conjugate consists of an anti-S. aureus antibody conjugated to a highly efficacious antibiotic that is activated only after it is released in the proteolytic environment of the phagolysosome. The antibody-antibiotic conjugate is superior to vancomycin for treatment of bacteraemia and provides direct evidence that intracellular S. aureus represents an important component of invasive infections.


Assuntos
Antibacterianos/farmacologia , Bacteriemia , Imunoconjugados/farmacologia , Imunoconjugados/uso terapêutico , Espaço Intracelular/microbiologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/efeitos dos fármacos , Vancomicina/farmacologia , Animais , Antibacterianos/uso terapêutico , Bacteriemia/tratamento farmacológico , Bacteriemia/microbiologia , Portador Sadio/tratamento farmacológico , Portador Sadio/microbiologia , Desenho de Fármacos , Feminino , Imunoconjugados/química , Espaço Intracelular/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina/patogenicidade , Camundongos , Testes de Sensibilidade Microbiana , Fagossomos/efeitos dos fármacos , Fagossomos/metabolismo , Fagossomos/microbiologia , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/patologia , Staphylococcus aureus/patogenicidade , Vancomicina/uso terapêutico
11.
Am J Physiol Endocrinol Metab ; 309(10): E829-39, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26394662

RESUMO

Nε-(carboxymethyl) lysine-conjugated bovine serum albumin (CML-BSA) is a major component of advanced glycation end products (AGEs). We hypothesised that AGEs reduce insulin secretion from pancreatic ß-cells by damaging mitochondrial functions and inducing mitophagy. Mitochondrial morphology and the occurrence of autophagy were examined in pancreatic islets of diabetic db/db mice and in the cultured CML-BSA-treated insulinoma cell line RIN-m5F. In addition, the effects of α-lipoic acid (ALA) on mitochondria in AGE-damaged tissues were evaluated. The diabetic db/db mouse exhibited an increase in the number of autophagosomes in damaged mitochondria and receptor for AGEs (RAGE). Treatment of db/db mice with ALA for 12 wk increased the number of mitochondria with well-organized cristae and fewer autophagosomes. Treatment of RIN-m5F cells with CML-BSA increased the level of RAGE protein and autophagosome formation, caused mitochondrial dysfunction, and decreased insulin secretion. CML-BSA also reduced mitochondrial membrane potential and ATP production, increased ROS and lipid peroxide production, and caused mitochondrial DNA deletions. Elevated fission protein dynamin-related protein 1 (Drp1) level and mitochondrial fragmentation demonstrated the unbalance of mitochondrial fusion and fission in CML-BSA-treated cells. Additionally, increased levels of Parkin and PTEN-induced putative kinase 1 protein suggest that fragmented mitochondria were associated with increased mitophagic activity, and ALA attenuated the CML-BSA-induced mitophage formation. Our study demonstrated that CML-BSA induced mitochondrial dysfunction and mitophagy in pancreatic ß-cells. The findings from this study suggest that increased concentration of AGEs may damage ß-cells and reduce insulin secretion.


Assuntos
Diabetes Mellitus/metabolismo , Produtos Finais de Glicação Avançada/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Lisina/análogos & derivados , Dinâmica Mitocondrial , Mitofagia , Animais , Antioxidantes/metabolismo , Antioxidantes/uso terapêutico , Biomarcadores/metabolismo , Linhagem Celular Tumoral , Diabetes Mellitus/dietoterapia , Diabetes Mellitus/patologia , Suplementos Nutricionais , Regulação para Baixo/efeitos dos fármacos , Produtos Finais de Glicação Avançada/antagonistas & inibidores , Produtos Finais de Glicação Avançada/farmacologia , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/ultraestrutura , Lisina/antagonistas & inibidores , Lisina/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Dinâmica Mitocondrial/efeitos dos fármacos , Mitofagia/efeitos dos fármacos , Oxidantes/antagonistas & inibidores , Oxidantes/farmacologia , Fagossomos/efeitos dos fármacos , Fagossomos/metabolismo , Fagossomos/ultraestrutura , Ratos , Receptor para Produtos Finais de Glicação Avançada/agonistas , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Soroalbumina Bovina/antagonistas & inibidores , Soroalbumina Bovina/farmacologia , Ácido Tióctico/metabolismo , Ácido Tióctico/uso terapêutico
12.
Sci Rep ; 5: 10903, 2015 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-26045184

RESUMO

Gaucher disease (GD) is caused by mutations in the GBA1 gene, which encodes lysosomal ß-glucocerebrosidase. Homozygosity for the L444P mutation in GBA1 is associated with high risk of neurological manifestations which are not improved by enzyme replacement therapy. Alternatively, pharmacological chaperones (PCs) capable of restoring the correct folding and trafficking of the mutant enzyme represent promising alternative therapies.Here, we report on how the L444P mutation affects mitochondrial function in primary fibroblast derived from GD patients. Mitochondrial dysfunction was associated with reduced mitochondrial membrane potential, increased reactive oxygen species (ROS), mitophagy activation and impaired autophagic flux.Both abnormalities, mitochondrial dysfunction and deficient ß-glucocerebrosidase activity, were partially restored by supplementation with coenzyme Q10 (CoQ) or a L-idonojirimycin derivative, N-[N'-(4-adamantan-1-ylcarboxamidobutyl)thiocarbamoyl]-1,6-anhydro-L-idonojirimycin (NAdBT-AIJ), and more markedly by the combination of both treatments. These data suggest that targeting both mitochondria function by CoQ and protein misfolding by PCs can be promising therapies in neurological forms of GD.


Assuntos
Inibidores Enzimáticos/farmacologia , Doença de Gaucher/metabolismo , Glucosilceramidase/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Ubiquinona/análogos & derivados , Autofagia/efeitos dos fármacos , Autofagia/genética , Biomarcadores , Ativação Enzimática , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Doença de Gaucher/tratamento farmacológico , Doença de Gaucher/genética , Expressão Gênica , Glucosilceramidase/genética , Humanos , Mutação , Fagossomos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Ubiquinona/farmacologia
13.
J Cell Mol Med ; 19(5): 1055-64, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25704599

RESUMO

We aim to investigate the effect of aurantiamide acetate isolated from the aerial parts of Clematis terniflora DC against gliomas. Human malignant glioma U87 and U251 cells were incubated with different concentrations (0-100 µM) of aurantiamide acetate. Aurantiamide acetate greatly decreased the cell viability in a dose- and time-dependent manner. It induced moderate mitochondrial fragmentation and the loss of mitochondrial membrane potential. No significant difference was found in the alternation of other intracellular organelles, although F-actin structure was slightly disturbed. Apparent ultrastructure alternation with increased autophagosome and autolysosome accumulation was observed in aurantiamide acetate-treated cells. The expression of LC3-II was greatly up-regulated in cells exposed to aurantiamide acetate (P < 0.05 compared with control). The cytoplasmic accumulation of autophagosomes and autolysosomes induced by aurantiamide acetate treatment was confirmed by fluorescent reporter protein labelling. Administration of chloroquine (CQ), which inhibits the fusion step of autophagosomes, further increased the accumulation of autophagosomes in the cytoplasm of U87 cells. Autophagy inhibition by 3-methyladenine, Bafilomycin A1 or CQ had no influence on aurantiamide acetate-induced cytotoxicity, whereas autophagy stimulator rapamycin significantly suppressed aurantiamide acetate-induced cell death. The anti-tumour effects of aurantiamide acetate were further evaluated in tumour-bearing nude mice. Intratumoural injection of aurantiamide acetate obviously suppressed tumour growth, and increased number of autophagic vacuoles was observed in tumour tissues of animals receiving aurantiamide acetate. Our findings suggest that aurantiamide acetate may suppress the growth of malignant gliomas by blocking autophagic flux.


Assuntos
Autofagia/efeitos dos fármacos , Neoplasias Encefálicas/prevenção & controle , Proliferação de Células/efeitos dos fármacos , Clematis/química , Dipeptídeos/farmacologia , Glioma/prevenção & controle , Animais , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Dipeptídeos/química , Dipeptídeos/isolamento & purificação , Relação Dose-Resposta a Droga , Glioma/metabolismo , Glioma/patologia , Células Hep G2 , Humanos , Camundongos Nus , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Estrutura Molecular , Células PC12 , Fagossomos/efeitos dos fármacos , Fagossomos/metabolismo , Fagossomos/ultraestrutura , Fitoterapia , Componentes Aéreos da Planta/química , Ratos , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Cell Death Dis ; 6: e1604, 2015 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-25611379

RESUMO

Osteosarcoma is the most common primary malignant tumor of bone, the long-term survival of which has stagnated in the past several decades. Celastrol, a triterpene from traditional Chinese medicine, has been proved to possess potent anti-tumor effect on various cancers. However, the effect of celastrol on human osteosarcoma and the underlying mechanisms remains to be elucidated. We reported here that celastrol could inhibit cell proliferation by causing G2/M phase arrest. Exposure to celastrol resulted in the activation of caspase-3, -8, and -9, indicating that celastrol induced apoptosis through both extrinsic and intrinsic pathways. Autophagy occurred in celastrol-treated cells as evidenced by formation of autophagosome and accumulation of LC3B-II. The celastrol-induced cell death was remarkably restored by the combination of autophagy and apoptosis inhibitors. Furthermore, inhibition of apoptosis enhanced autophagy while suppression of autophagy diminished apoptosis. Celastrol also induced JNK activation and ROS generation. The JNK inhibitor significantly attenuated celastrol-triggered apoptosis and autophagy while ROS scavenger could completely reverse them. The ROS scavenger also prevented G2/M phase arrest and phosphorylation of JNK. Importantly, we found that celastrol had the similar effects on primary osteosarcoma cells. Finally, in vivo, celastrol suppressed tumor growth in the mouse xenograft model. Taken together, our results revealed that celastrol caused G2/M phase arrest, induced apoptosis and autophagy via the ROS/JNK signaling pathway in human osteosarcoma cells. Celastrol is therefore a promising candidate for development of antitumor drugs targeting osteosarcoma.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Osteossarcoma/enzimologia , Osteossarcoma/patologia , Espécies Reativas de Oxigênio/metabolismo , Triterpenos/farmacologia , Animais , Caspases/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Modelos Biológicos , Osteossarcoma/ultraestrutura , Triterpenos Pentacíclicos , Fagossomos/efeitos dos fármacos , Fagossomos/metabolismo , Fagossomos/ultraestrutura , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Mol Cell Proteomics ; 14(4): 870-81, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25616868

RESUMO

Upon entry into mammalian host cells, the pathogenic bacterium Francisella must import host cell arginine to multiply actively in the host cytoplasm. We identified and functionally characterized an arginine transporter (hereafter designated ArgP) whose inactivation considerably delayed bacterial phagosomal escape and intracellular multiplication. Intramacrophagic growth of the ΔargP mutant was fully restored upon supplementation of the growth medium with excess arginine, in both F. tularensis subsp. novicida and F. tularensis subsp. holarctica LVS, demonstrating the importance of arginine acquisition in these two subspecies. High-resolution mass spectrometry revealed that arginine limitation reduced the amount of most of the ribosomal proteins in the ΔargP mutant. In response to stresses such as nutritional limitation, repression of ribosomal protein synthesis has been observed in all kingdoms of life. Arginine availability may thus contribute to the sensing of the intracellular stage of the pathogen and to trigger phagosomal egress. All MS data have been deposited in the ProteomeXchange database with identifier PXD001584 (http://proteomecentral.proteomexchange.org/dataset/PXD001584).


Assuntos
Arginina/metabolismo , Francisella/metabolismo , Interações Hospedeiro-Patógeno , Fagossomos/microbiologia , Proteínas Ribossômicas/metabolismo , Animais , Autofagia , Proteínas de Bactérias/metabolismo , Vacinas Bacterianas/imunologia , Análise por Conglomerados , Citosol/metabolismo , Feminino , Francisella/patogenicidade , Macrófagos/metabolismo , Macrófagos/microbiologia , Macrófagos/ultraestrutura , Proteínas de Membrana Transportadoras/metabolismo , Camundongos Endogâmicos BALB C , Viabilidade Microbiana , Modelos Biológicos , Mutação/genética , Fagossomos/metabolismo , Fagossomos/ultraestrutura , Transporte Proteico , Proteoma/metabolismo , Estresse Fisiológico , Frações Subcelulares/metabolismo , Virulência
16.
PLoS One ; 9(11): e113682, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25419658

RESUMO

Bergamot (Citrus bergamia, Risso et Poiteau) essential oil (BEO) is a well characterized, widely used plant extract. BEO exerts anxiolytic, analgesic and neuroprotective activities in rodents through mechanisms that are only partly known and need to be further investigated. To gain more insight into the biological effects of this essential oil, we tested the ability of BEO (0.005-0.03%) to modulate autophagic pathways in human SH-SY5Y neuroblastoma cells. BEO-treated cells show increased LC3II levels and appearance of dot-like formations of endogenous LC3 protein that colocalize with the lysosome marker LAMP-1. Autophagic flux assay using bafilomycin A1 and degradation of the specific autophagy substrate p62 confirmed that the observed increase of LC3II levels in BEO-exposed cells is due to autophagy induction rather than to a decreased autophagosomal turnover. Induction of autophagy is an early and not cell-line specific response to BEO. Beside basal autophagy, BEO also enhanced autophagy triggered by serum starvation and rapamycin indicating that the underlying mechanism is mTOR independent. Accordingly, BEO did not affect the phosphorylation of ULK1 (Ser757) and p70(S6K) (Thr389), two downstream targets of mTOR. Furthermore, induction of autophagy by BEO is beclin-1 independent, occurs in a concentration-dependent manner and is unrelated to the ability of BEO to induce cell death. In order to identify the active constituents responsible for these effects, the two most abundant monoterpenes found in the essential oil, d-limonene (125-750 µM) and linalyl acetate (62.5-375 µM), were individually tested at concentrations comparable to those found in 0.005-0.03% BEO. The same features of stimulated autophagy elicited by BEO were reproduced by D-limonene, which rapidly increases LC3II and reduces p62 levels in a concentration-dependent manner. Linalyl acetate was ineffective in replicating BEO effects; however, it greatly enhanced LC3 lipidation triggered by D-limonene.


Assuntos
Autofagia/efeitos dos fármacos , Cicloexenos/farmacologia , Óleos Voláteis/farmacologia , Óleos de Plantas/farmacologia , Terpenos/farmacologia , Western Blotting , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Limoneno , Lisossomos/metabolismo , Células MCF-7 , Fusão de Membrana/efeitos dos fármacos , Microscopia Confocal , Proteínas Associadas aos Microtúbulos/metabolismo , Monoterpenos/farmacologia , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Fagossomos/metabolismo
17.
J Biol Chem ; 289(48): 33568-89, 2014 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-25301941

RESUMO

Autophagy functions as a survival mechanism during cellular stress and contributes to resistance against anticancer agents. The selective antitumor and antimetastatic chelator di-2-pyridylketone 4,4-dimethyl-3-thiosemicarbazone (Dp44mT) causes lysosomal membrane permeabilization and cell death. Considering the integral role of lysosomes in autophagy and cell death, it was important to assess the effect of Dp44mT on autophagy to further understand its mechanism of action. Notably, Dp44mT affected autophagy by two mechanisms. First, concurrent with its antiproliferative activity, Dp44mT increased the expression of the classical autophagic marker LC3-II as a result of induced autophagosome synthesis. Second, this effect was supplemented by a reduction in autophagosome degradation as shown by the accumulation of the autophagic substrate and receptor p62. Conversely, the classical iron chelator desferrioxamine induced autophagosome accumulation only by inhibiting autophagosome degradation. The formation of redox-active iron or copper Dp44mT complexes was critical for its dual effect on autophagy. The cytoprotective antioxidant N-acetylcysteine inhibited Dp44mT-induced autophagosome synthesis and p62 accumulation. Importantly, Dp44mT inhibited autophagosome degradation via lysosomal disruption. This effect prevented the fusion of lysosomes with autophagosomes to form autolysosomes, which is crucial for the completion of the autophagic process. The antiproliferative activity of Dp44mT was suppressed by Beclin1 and ATG5 silencing, indicating the role of persistent autophagosome synthesis in Dp44mT-induced cell death. These studies demonstrate that Dp44mT can overcome the prosurvival activity of autophagy in cancer cells by utilizing this process to potentiate cell death.


Assuntos
Antineoplásicos/farmacologia , Autofagia/efeitos dos fármacos , Lisossomos/metabolismo , Neoplasias/tratamento farmacológico , Fagossomos/metabolismo , Tiossemicarbazonas/farmacologia , Acetilcisteína/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cobre/metabolismo , Desferroxamina/farmacologia , Feminino , Sequestradores de Radicais Livres/farmacologia , Humanos , Ferro/metabolismo , Lisossomos/patologia , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Oxirredução/efeitos dos fármacos , Fagossomos/patologia , Sideróforos/farmacologia
18.
J Biol Chem ; 289(34): 23938-50, 2014 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-25023287

RESUMO

Autophagy is one of the major degradation pathways for cytoplasmic components. The autophagic isolation membrane is a unique membrane whose content of unsaturated fatty acids is very high. However, the molecular mechanisms underlying formation of this membrane, including the roles of unsaturated fatty acids, remain to be elucidated. From a chemical library consisting of structurally diverse compounds, we screened for novel inhibitors of starvation-induced autophagy by measuring LC3 puncta formation in mouse embryonic fibroblasts stably expressing GFP-LC3. One of the inhibitors we identified, 2,5-pyridinedicarboxamide, N2,N5-bis[5-[(dimethylamino)carbonyl]-4-methyl-2-thiazolyl], has a molecular structure similar to that of a known stearoyl-CoA desaturase (SCD) 1 inhibitor. To determine whether SCD1 inhibition influences autophagy, we examined the effects of the SCD1 inhibitor 28c. This compound strongly inhibited starvation-induced autophagy, as determined by LC3 puncta formation, immunoblot analyses of LC3, electron microscopic observations, and p62/SQSTM1 accumulation. Overexpression of SCD1 or supplementation with oleic acid, which is a catalytic product of SCD1 abolished the inhibition of autophagy by 28c. Furthermore, 28c suppressed starvation-induced autophagy without affecting mammalian target of rapamycin activity, and also inhibited rapamycin-induced autophagy. In addition to inhibiting formation of LC3 puncta, 28c also inhibited formation of ULK1, WIPI1, Atg16L, and p62/SQSTM1 puncta. These results suggest that SCD1 activity is required for the earliest step of autophagosome formation.


Assuntos
Autofagia , Fagossomos/metabolismo , Estearoil-CoA Dessaturase/metabolismo , Animais , Sequência de Bases , Células Cultivadas , Primers do DNA , Ácidos Graxos/metabolismo , Camundongos , Microscopia de Fluorescência , Transporte Proteico , Reação em Cadeia da Polimerase em Tempo Real
19.
Cell Cycle ; 13(12): 1987-94, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24769862

RESUMO

Epidemiological studies and clinical trials revealed that chronic consumption coffee is associated with the inhibition of several metabolic diseases as well as reduction in overall and cause-specific mortality. We show that both natural and decaffeinated brands of coffee similarly rapidly trigger autophagy in mice. One to 4 h after coffee consumption, we observed an increase in autophagic flux in all investigated organs (liver, muscle, heart) in vivo, as indicated by the increased lipidation of LC3B and the reduction of the abundance of the autophagic substrate sequestosome 1 (p62/SQSTM1). These changes were accompanied by the inhibition of the enzymatic activity of mammalian target of rapamycin complex 1 (mTORC1), leading to the reduced phosphorylation of p70(S6K), as well as by the global deacetylation of cellular proteins detectable by immunoblot. Immunohistochemical analyses of transgenic mice expressing a GFP-LC3B fusion protein confirmed the coffee-induced relocation of LC3B to autophagosomes, as well as general protein deacetylation. Altogether, these results indicate that coffee triggers 2 phenomena that are also induced by nutrient depletion, namely a reduction of protein acetylation coupled to an increase in autophagy. We speculate that polyphenols contained in coffee promote health by stimulating autophagy.


Assuntos
Autofagia , Café/metabolismo , Acetilação , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Feminino , Fígado/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos Endogâmicos C57BL , Complexos Multiproteicos/metabolismo , Músculo Esquelético/metabolismo , Miocárdio/metabolismo , Fagossomos/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Fator de Transcrição TFIIH , Fatores de Transcrição/metabolismo
20.
Cell Death Dis ; 5: e1193, 2014 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-24763050

RESUMO

The growing number of studies suggested that inhibition of autophagy enhances the efficacy of Akt kinase inhibitors in cancer therapy. Here, we provide evidence that ML-9, a widely used inhibitor of Akt kinase, myosin light-chain kinase (MLCK) and stromal interaction molecule 1 (STIM1), represents the 'two-in-one' compound that stimulates autophagosome formation (by downregulating Akt/mammalian target of rapamycin (mTOR) pathway) and inhibits their degradation (by acting like a lysosomotropic agent and increasing lysosomal pH). We show that ML-9 as a monotherapy effectively induces prostate cancer cell death associated with the accumulation of autophagic vacuoles. Further, ML-9 enhances the anticancer activity of docetaxel, suggesting its potential application as an adjuvant to existing anticancer chemotherapy. Altogether, our results revealed the complex effect of ML-9 on autophagy and indentified ML-9 as an attractive tool for targeting autophagy in cancer therapy through dual inhibition of both the Akt pathway and the autophagy.


Assuntos
Autofagia/efeitos dos fármacos , Azepinas/farmacologia , Lisossomos/efeitos dos fármacos , Neoplasias da Próstata/patologia , Cálcio/metabolismo , Linhagem Celular Tumoral , Classe III de Fosfatidilinositol 3-Quinases/metabolismo , Regulação para Baixo/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Humanos , Concentração de Íons de Hidrogênio/efeitos dos fármacos , Lisossomos/ultraestrutura , Masculino , Modelos Biológicos , Fagossomos/efeitos dos fármacos , Fagossomos/metabolismo , Fagossomos/ultraestrutura , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/ultraestrutura , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA