Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 197
Filtrar
1.
Biomater Sci ; 11(5): 1810-1827, 2023 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-36655818

RESUMO

Stimuli-responsive cross-linked nanocarriers that can induce lysosomal cell death (LCD) via lysosomal membrane permeabilization (LMP) represent a new class of delivery platforms and have attracted the attention of researchers in the biomedical field. The advantages of such cross-linked nanocarriers are as follows (i) they remain intact during blood circulation; and (ii) they reach the target site via specific receptor-mediated endocytosis leading to the enhancement of therapeutic efficacy and reduction of side effects. Herein, we have synthesized a mannose-6-phosphate (M6P) based amphiphilic ABC type tri-block copolymer having two chains of FDA-approved poly(ε-caprolactone) (PCL) as the hydrophobic block, and poly(S-(o-nitrobenzyl)-L-cysteine) (NBC) acts as the photoresponsive crosslinker block. Two different tri-block copolymers, [(PCL35)2-b-NBC20-b-M6PGP20] and [(PCL35)2-b-NBC15-b-M6PGP20], were synthesized which upon successful self-assembly initially formed spherical uncross-linked "micellar-type" aggregates (UCL-M) and vesicles (UCL-V), respectively. The uncross-linked nanocarriers upon UV treatment for thirty minutes were covalently crosslinked in the middle PNBC block giving rise to the di-sulfide bonds and forming interface cross-linked "micellar-type" aggregates (ICL-M) and vesicles (ICL-V). DLS, TEM, and AFM techniques were used to successfully characterize the morphology of these nanocarriers. The dual stimuli (redox and enzyme) responsiveness of the cross-linked nanocarriers and their trafficking to the lysosome in mammalian cells via receptor-mediated endocytosis was probed using confocal microscopy images. Furthermore, the addition of a chloroquine (CQ, a known lysosomotropic agent) encapsulated cross-linked nanocarrier (CQ@ICL-V) to non-cancerous (HEK-293T) cells and liver (HepG2), and breast cancer cells (MDA-MB-231) was found to initiate lysosomal membrane permeabilization (LMP) followed by lysosomal destabilization which eventually led to lysosomal cell death (LCD). Due to the targeted delivery of CQ to the lysosomes of cancerous cells, almost a 90% smaller amount of CQ was able to achieve similar cell death to CQ alone.


Assuntos
Manosefosfatos , Polímeros , Animais , Polímeros/química , Manosefosfatos/metabolismo , Micelas , Lisossomos/metabolismo , Mamíferos
2.
Commun Biol ; 6(1): 48, 2023 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-36639722

RESUMO

N-glycosylation is implicated in cancers and aberrant N-glycosylation is recognized as a hallmark of cancer. Here, we mapped and compared the site-specific N-glycoproteomes of colon cancer HCT116 cells and isogenic non-tumorigenic DNMT1/3b double knockout (DKO1) cells using Fbs1-GYR N-glycopeptide enrichment technology and trapped ion mobility spectrometry. Many significant changes in site-specific N-glycosylation were revealed, providing a molecular basis for further elucidation of the role of N-glycosylation in protein function. HCT116 cells display hypersialylation especially in cell surface membrane proteins. Both HCT116 and DKO1 show an abundance of paucimannose and 80% of paucimannose-rich proteins are annotated to reside in exosomes. The most striking N-glycosylation alteration was the degree of mannose-6-phosphate (M6P) modification. N-glycoproteomic analyses revealed that HCT116 displays hyper-M6P modification, which was orthogonally validated by M6P immunodetection. Significant observed differences in N-glycosylation patterns of the major M6P receptor, CI-MPR in HCT116 and DKO1 may contribute to the hyper-M6P phenotype of HCT116 cells. This comparative site-specific N-glycoproteome analysis provides a pool of potential N-glycosylation-related cancer biomarkers, but also gives insights into the M6P pathway in cancer.


Assuntos
Manosefosfatos , Neoplasias , Humanos , Glicosilação , Manosefosfatos/química , Manosefosfatos/metabolismo , Neoplasias/genética
3.
Nat Commun ; 13(1): 5351, 2022 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-36096887

RESUMO

The mannose-6-phosphate (M6P) biosynthetic pathway for lysosome biogenesis has been studied for decades and is considered a well-understood topic. However, whether this pathway is regulated remains an open question. In a genome-wide CRISPR/Cas9 knockout screen, we discover TMEM251 as the first regulator of the M6P modification. Deleting TMEM251 causes mistargeting of most lysosomal enzymes due to their loss of M6P modification and accumulation of numerous undigested materials. We further demonstrate that TMEM251 localizes to the Golgi and is required for the cleavage and activity of GNPT, the enzyme that catalyzes M6P modification. In zebrafish, TMEM251 deletion leads to severe developmental defects including heart edema and skeletal dysplasia, which phenocopies Mucolipidosis Type II. Our discovery provides a mechanism for the newly discovered human disease caused by TMEM251 mutations. We name TMEM251 as GNPTAB cleavage and activity factor (GCAF) and its related disease as Mucolipidosis Type V.


Assuntos
Proteínas de Membrana , Mucolipidoses , Peixe-Zebra , Animais , Humanos , Lisossomos/metabolismo , Manosefosfatos/metabolismo , Proteínas de Membrana/metabolismo , Mucolipidoses/genética , Mucolipidoses/metabolismo , Transferases (Outros Grupos de Fosfato Substituídos)/genética , Transferases (Outros Grupos de Fosfato Substituídos)/metabolismo , Peixe-Zebra/metabolismo
4.
Chembiochem ; 22(2): 434-440, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-32864819

RESUMO

Mannose-6-phosphate (M6P) is recognized by the mannose-6-phosphate receptor and plays an important role in the transport of cargo to the endosomes, making it an attractive tool to improve endosomal trafficking of vaccines. We describe herein the assembly of peptide antigen conjugates carrying clusters of mannose-6-C-phosphonates (M6Po). The M6Po's are stable M6P mimics that are resistant to cleavage of the phosphate group by endogenous phosphatases. Two different strategies for the incorporation of the M6Po clusters in the conjugate have been developed: the first relies on a "post-assembly" click approach employing an M6Po bearing an alkyne functionality; the second hinges on an M6Po C-glycoside amino acid building block that can be used in solid-phase peptide synthesis. The generated conjugates were further equipped with a TLR7 ligand to stimulate dendritic cell (DC) maturation. While antigen presentation is hindered by the presence of the M6Po clusters, the incorporation of the M6Po clusters leads to increased activation of DCs, thus demonstrating their potential in improving vaccine adjuvanticity by intraendosomally active TLR ligands.


Assuntos
Antígenos/metabolismo , Manosefosfatos/metabolismo , Peptídeos/metabolismo , Receptores Toll-Like/metabolismo , Antígenos/química , Humanos , Ligantes , Manosefosfatos/química , Estrutura Molecular , Peptídeos/química , Receptores Toll-Like/química
5.
Nat Commun ; 11(1): 6343, 2020 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-33311467

RESUMO

D-mannose is a monosaccharide approximately a hundred times less abundant than glucose in human blood. Previous studies demonstrated that supraphysiological levels of D-mannose inhibit tumour growth and stimulate regulatory T cell differentiation. It is not known whether D-mannose metabolism affects the function of non-proliferative cells, such as inflammatory macrophages. Here, we show that D-mannose suppresses LPS-induced macrophage activation by impairing IL-1ß production. In vivo, mannose administration improves survival in a mouse model of LPS-induced endotoxemia as well as decreases progression in a mouse model of DSS-induced colitis. Phosphomannose isomerase controls response of LPS-activated macrophages to D-mannose, which impairs glucose metabolism by raising intracellular mannose-6-phosphate levels. Such alterations result in the suppression of succinate-mediated HIF-1α activation, imposing a consequent reduction of LPS-induced Il1b expression. Disclosing an unrecognized metabolic hijack of macrophage activation, our study points towards safe D-mannose utilization as an effective intervention against inflammatory conditions.


Assuntos
Interleucina-1beta/metabolismo , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Manose/metabolismo , Manose/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Colite/metabolismo , Colite/patologia , Regulação da Expressão Gênica , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Inflamação/metabolismo , Interleucina-1beta/genética , Lipopolissacarídeos/efeitos adversos , Manosefosfatos/metabolismo , Redes e Vias Metabólicas/efeitos dos fármacos , Metabolômica , Monócitos/metabolismo
6.
Int J Mol Sci ; 21(18)2020 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-32957425

RESUMO

Mucolipidosis II and III (ML II/III) are caused by a deficiency of uridine-diphosphate N-acetylglucosamine: lysosomal-enzyme-N-acetylglucosamine-1-phosphotransferase (GlcNAc-1-phosphotransferase, EC2.7.8.17), which tags lysosomal enzymes with a mannose 6-phosphate (M6P) marker for transport to the lysosome. The process is performed by a sequential two-step process: first, GlcNAc-1-phosphotransferase catalyzes the transfer of GlcNAc-1-phosphate to the selected mannose residues on lysosomal enzymes in the cis-Golgi network. The second step removes GlcNAc from lysosomal enzymes by N-acetylglucosamine-1-phosphodiester α-N-acetylglucosaminidase (uncovering enzyme) and exposes the mannose 6-phosphate (M6P) residues in the trans-Golgi network, in which the enzymes are targeted to the lysosomes by M6Preceptors. A deficiency of GlcNAc-1-phosphotransferase causes the hypersecretion of lysosomal enzymes out of cells, resulting in a shortage of multiple lysosomal enzymes within lysosomes. Due to a lack of GlcNAc-1-phosphotransferase, the accumulation of cholesterol, phospholipids, glycosaminoglycans (GAGs), and other undegraded substrates occurs in the lysosomes. Clinically, ML II and ML III exhibit quite similar manifestations to mucopolysaccharidoses (MPSs), including specific skeletal deformities known as dysostosis multiplex and gingival hyperplasia. The life expectancy is less than 10 years in the severe type, and there is no definitive treatment for this disease. In this review, we have described the updated diagnosis and therapy on ML II/III.


Assuntos
Terapia de Reposição de Enzimas/métodos , Terapia Genética/métodos , Transplante de Células-Tronco Hematopoéticas/métodos , Lisossomos/metabolismo , Mucolipidoses/diagnóstico , Animais , Transporte Biológico Ativo , Modelos Animais de Doenças , Glicosaminoglicanos/metabolismo , Humanos , Manosefosfatos/metabolismo , Mucolipidoses/enzimologia , Mucolipidoses/fisiopatologia , Mucolipidoses/terapia
7.
Protein Expr Purif ; 170: 105589, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32027983

RESUMO

The cation-independent mannose-6-phosphate receptor (CI-M6PR, aka insulin-like growth factor II receptor or IGFIIR) is a membrane protein that plays a central role in the trafficking of lysosomal acid hydrolases into lysosomes via mannose-6-phosphate (M6P) binding domains. In order to maintain cellular metabolic/catabolic homeostasis, newly synthesized lysosomal acid hydrolases are required to bind to M6PR for transit. Acid hydrolases secreted by cells can also be internalized via M6PR residing on the cell membrane and are transported to the lysosomes, a feature that enables enzyme replacement therapy for the treatment of several lysosomal storage disorders. Therefore, a thorough characterization of this receptor is critical to the development of lysosomal enzyme-based therapeutics that utilize M6PR for drug delivery to the lysosome. However, the extracellular domain (ECD) of M6PR is highly complex, containing 15-mannose receptor homology (MRH) domains. In addition, homodimerization of the receptor can occur at the membrane, making its characterization challenging. In this study, a novel human M6PR (hM6PR)-overexpressing cell line originally established for hM6PR cellular uptake assay was utilized for production of hM6PR-ECD, and a novel small molecule biomimetic (aminophenyl-M6P) affinity resin was developed for the purification of M6PR-ECD. The affinity-purified hM6PR-ECD was monomeric, contained 14 intact MRH domains (1-14) and a partial MRH domain 15, and was successfully employed in ELISA-based and surface plasmon resonance-based binding assays to demonstrate its ligand-binding functionality, making it suitable for the evaluation of biotherapeutics that utilize M6PR for cellular internalization.


Assuntos
Aminofenóis/química , Materiais Biomiméticos/química , Membrana Celular/enzimologia , Manosefosfatos/química , Receptor IGF Tipo 2/isolamento & purificação , Sequência de Aminoácidos , Aminofenóis/metabolismo , Materiais Biomiméticos/metabolismo , Linhagem Celular Tumoral , Membrana Celular/química , Cromatografia de Afinidade , Ensaios Enzimáticos , Ensaio de Imunoadsorção Enzimática , Fibroblastos/química , Fibroblastos/enzimologia , Expressão Gênica , Humanos , Cinética , Manosefosfatos/metabolismo , Domínios Proteicos , Receptor IGF Tipo 2/genética , Receptor IGF Tipo 2/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Ressonância de Plasmônio de Superfície
8.
J Virol ; 93(5)2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30541844

RESUMO

Human herpesvirus 8 (HHV-8) viral interleukin-6 (vIL-6) localizes largely to the endoplasmic reticulum (ER) and here associates functionally with both the gp130 signal transducer and the novel ER membrane protein vitamin K epoxide reductase complex subunit 1 variant-2 (VKORC1v2). The latter interaction contributes to the viability of latently infected primary effusion lymphoma (PEL) cells and to HHV-8 productive replication, in part via promotion of ER-associated degradation (ERAD) of nascent pro-cathepsin D (pCatD) and consequent suppression of lysosome-localized proapoptotic mature CatD. Here we report that VKORC1v2 associates with insulin-like growth factor 2 receptor (IGF2R), also known as cation-independent mannose-6-phosphate receptor, which is involved in trafficking of mannose-6-phosphate-conjugated glycoproteins to lysosomes. VKORC1v2 effected reduced IGF2R expression in a manner dependent on VKORC1v2-IGF2R interaction, while vIL-6, which could inhibit VKORC1v2-IGF2R interaction, effected increased expression of IGF2R. These effects were independent of changes in IGF2R mRNA levels, indicating likely posttranslational mechanisms. In kinetic analyses involving labeling of either newly synthesized or preexisting IGF2R, vIL-6 promoted accumulation of the former while having no detectable effect on the latter. Furthermore, vIL-6 led to decreased K48-linked ubiquitination of IGF2R and suppression of ERAD proteins effected increased IGF2R expression and loss of IGF2R regulation by vIL-6. Depletion-based experiments identified IGF2R as a promoter of PEL cell viability and virus yields from lytically reactivated cultures. Our findings identify ER-transiting nascent IGF2R as an interaction partner of VKORC1v2 and target of vIL-6 regulation and IGF2R as a positive contributor to HHV-8 biology, thereby extending understanding of the mechanisms of VKORC1v2-associated vIL-6 function.IMPORTANCE HHV-8 vIL-6 promotes productive replication in the context of reactivated lytic replication in primary effusion lymphoma (PEL) and endothelial cells and sustains latently infected PEL cell viability. Viral IL-6 is also considered to contribute significantly to HHV-8-associated pathogenesis, since vIL-6 can promote cell proliferation, cell survival, and angiogenesis that are characteristic of HHV-8-associated Kaposi's sarcoma, PEL and multicentric Castleman's disease (MCD), in addition to proinflammatory activities observed in MCD-like "Kaposi's sarcoma-associated herpesvirus-induced cytokine syndrome." We show in the present study that vIL-6 can promote productive replication and latent PEL cell viability through upregulation of the mannose-6-phosphate- and peptide hormone-interacting receptor IGF2R, which is a positive factor in HHV-8 biology via these activities. VKORC1v2-enhanced ER-associated degradation of IGF2R and vIL-6 promotion of IGF2R expression through prevention of its interaction with VKORC1v2 and consequent rescue from degradation represent newly recognized activities of VKOCR1v2 and vIL-6.


Assuntos
Células Endoteliais/virologia , Herpesvirus Humano 8/metabolismo , Interleucina-6/metabolismo , Linfoma de Efusão Primária/virologia , Receptor IGF Tipo 2/metabolismo , Vitamina K Epóxido Redutases/metabolismo , Catepsina D/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/genética , Sobrevivência Celular/genética , Receptor gp130 de Citocina/metabolismo , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/virologia , Precursores Enzimáticos/metabolismo , Células HEK293 , Humanos , Manosefosfatos/metabolismo , Receptor IGF Tipo 2/biossíntese , Receptor IGF Tipo 2/genética , Ubiquitinação , Ativação Viral/genética , Latência Viral/genética , Replicação Viral/genética
9.
Nature ; 563(7733): 719-723, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30464341

RESUMO

It is now well established that tumours undergo changes in cellular metabolism1. As this can reveal tumour cell vulnerabilities and because many tumours exhibit enhanced glucose uptake2, we have been interested in how tumour cells respond to different forms of sugar. Here we report that the monosaccharide mannose causes growth retardation in several tumour types in vitro, and enhances cell death in response to major forms of chemotherapy. We then show that these effects also occur in vivo in mice following the oral administration of mannose, without significantly affecting the weight and health of the animals. Mechanistically, mannose is taken up by the same transporter(s) as glucose3 but accumulates as mannose-6-phosphate in cells, and this impairs the further metabolism of glucose in glycolysis, the tricarboxylic acid cycle, the pentose phosphate pathway and glycan synthesis. As a result, the administration of mannose in combination with conventional chemotherapy affects levels of anti-apoptotic proteins of the Bcl-2 family, leading to sensitization to cell death. Finally we show that susceptibility to mannose is dependent on the levels of phosphomannose isomerase (PMI). Cells with low levels of PMI are sensitive to mannose, whereas cells with high levels are resistant, but can be made sensitive by RNA-interference-mediated depletion of the enzyme. In addition, we use tissue microarrays to show that PMI levels also vary greatly between different patients and different tumour types, indicating that PMI levels could be used as a biomarker to direct the successful administration of mannose. We consider that the administration of mannose could be a simple, safe and selective therapy in the treatment of cancer, and could be applicable to multiple tumour types.


Assuntos
Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Manose/metabolismo , Manose/farmacologia , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Administração Oral , Animais , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais/metabolismo , Peso Corporal/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Sinergismo Farmacológico , Feminino , Glucose/metabolismo , Glicólise/efeitos dos fármacos , Humanos , Manose/administração & dosagem , Manose/uso terapêutico , Manose-6-Fosfato Isomerase/deficiência , Manose-6-Fosfato Isomerase/genética , Manose-6-Fosfato Isomerase/metabolismo , Manosefosfatos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Neoplasias/classificação , Neoplasias/patologia , Interferência de RNA , Proteína bcl-X/metabolismo
10.
Adv Healthc Mater ; 7(14): e1701398, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29719138

RESUMO

In order to overcome the main disadvantages of conventional cancer therapies, which prove to be inadequate because of their lack of selectivity, the development of targeted delivery systems is one of the main focuses in anticancer research. It is repeatedly shown that decorating the surface of nanocarriers with high-affinity targeting ligands, such as peptides or small molecules, is an effective way to selectively deliver therapeutics by enhancing their specific cellular uptake via the binding between a specific receptor and the nanosystems. Nowadays, the need of finding new potential biological targets with a high endocytic efficiency as well as a low tendency to mutate is urgent and, in this context, mannose and mannose-6-phosphate receptors appear promising to target anticancer drugs to cells where their expression is upregulated. Moreover, they open the path to encouraging applications in immune-based and gene therapies as well as in theragnostic purposes. In this work, the potential of mannose- and mannose-6-phosphate-targeted delivery systems in cancer therapy is discussed, emphasizing their broad application both in direct treatments against cancer cells with conventional chemotherapeutics or by gene therapy and also their encouraging capabilities in immunotherapy and diagnostics purposes.


Assuntos
Manose/metabolismo , Manosefosfatos/metabolismo , Receptor IGF Tipo 2/metabolismo , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/uso terapêutico , Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos/métodos , Humanos , Imunoterapia , Nanomedicina/métodos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/terapia
11.
Microbiology (Reading) ; 164(3): 369-382, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29458553

RESUMO

Polyprenol phosphate mannose (PPM) is a lipid-linked sugar donor used by extra-cytoplasmic glycosyl tranferases in bacteria. PPM is synthesized by polyprenol phosphate mannose synthase, Ppm1, and in most Actinobacteria is used as the sugar donor for protein O-mannosyl transferase, Pmt, in protein glycosylation. Ppm1 and Pmt have homologues in yeasts and humans, where they are required for protein O-mannosylation. Actinobacteria also use PPM for lipoglycan biosynthesis. Here we show that ppm1 mutants of Streptomyces coelicolor have increased susceptibility to a number of antibiotics that target cell wall biosynthesis. The pmt mutants also have mildly increased antibiotic susceptibilities, in particular to ß-lactams and vancomycin. Despite normal induction of the vancomycin gene cluster, vanSRJKHAX, the pmt and ppm1 mutants remained highly vancomycin sensitive indicating that the mechanism of resistance is blocked post-transcriptionally. Differential RNA expression analysis indicated that catabolic pathways were downregulated and anabolic ones upregulated in the ppm1 mutant compared to the parent or complemented strains. Of note was the increase in expression of fatty acid biosynthetic genes in the ppm1- mutant. A change in lipid composition was confirmed using Raman spectroscopy, which showed that the ppm1- mutant had a greater relative proportion of unsaturated fatty acids compared to the parent or the complemented mutant. Taken together, these data suggest that an inability to synthesize PPM (ppm1) and loss of the glycoproteome (pmt- mutant) can detrimentally affect membrane or cell envelope functions leading to loss of intrinsic and, in the case of vancomycin, acquired antibiotic resistance.


Assuntos
Antibacterianos/farmacologia , Farmacorresistência Bacteriana/efeitos dos fármacos , Manosiltransferases/deficiência , Manosiltransferases/genética , Streptomyces coelicolor/efeitos dos fármacos , Streptomyces coelicolor/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Parede Celular/efeitos dos fármacos , Farmacorresistência Bacteriana/genética , Ácidos Graxos Insaturados/química , Expressão Gênica , Perfilação da Expressão Gênica , Metabolismo dos Lipídeos , Manosefosfatos/metabolismo , Manosiltransferases/metabolismo , Testes de Sensibilidade Microbiana , Mutação , Análise Espectral Raman , Streptomyces coelicolor/enzimologia , Streptomyces coelicolor/metabolismo
12.
J Microbiol Biotechnol ; 27(2): 372-379, 2017 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-27817186

RESUMO

The transport of lysosomal enzymes into the lysosomes depends on the phosphorylation of their chains and the binding of the phosphorylated residues to mannose-6-phosphate receptors. The efficiency of separation depends more on the phosphodiesterases (PDEs) than on the activity of the phosphorylation of mannose residues and can be determined in vitro. PDEs play important roles in regulation of the activation of lysosomes. The expression of proteins was confirmed by western blotting. All PDE4 series protein expression was reduced in high concentrations of rolipram. As a result of observing the fluorescence intensity after rolipram treatment, the lysosomal enzyme was activated at low concentrations and suppressed at high concentrations. High concentrations of rolipram recovered the original function. Antimicrobial activity was not shown in either 10 or 100 µ concentrations of rolipram in treated HeLa cells in vitro. However, the higher anticancer activity at lower rolipram concentration was shown in lysosomal enzyme treated with 10 µ of rolipram. The anticancer activity was confirmed through cathepsin B and D assay. Tranfection allowed examination of the relationship between PDE4 and lysosomal activity in more detail. Protein expression was confirmed to be reduced. Fluorescence intensity showed decreased activity of lysosomes and ROS in cells transfected with the antisense sequences of PDE4 A, B, C, and D. PDE4A showed anticancer activity, whereas lysosome from cells transfected with the antisense sequences of PDE4 B, C, and D had decreased anticancer activity. These results showed the PDE4 A, B, C, and D are conjunctly related with lysosomal activity.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/genética , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Regulação da Expressão Gênica , Lisossomos/enzimologia , Lisossomos/fisiologia , Animais , Western Blotting , Catepsina B/análise , AMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/química , Células HeLa , Humanos , Lisossomos/efeitos dos fármacos , Manosefosfatos/metabolismo , Fosforilação , Rolipram/farmacologia , Transfecção
13.
J Drug Target ; 25(4): 342-349, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27822974

RESUMO

The epithelial barrier in the respiratory system is a major obstacle for drug delivery to the systemic circulation in the lung. Epithelial barrier hinders the transport of large macromolecules or polar drugs. Essential components of this epithelial fence are physical intercellular structures termed tight junctions. Therefore, modulating tight junctions can enhance paracellular transport across epithelial barrier. In this study, the effect of some of non-specific tight junction modulators (TJMs); (Sodium (Na) decanoate, oleic acid and ethyleneglycol-bis-(ß-aminoethyl ether)-N, N'-tetraacetic acid (EGTA)) with established effect on intestinal tight junctions was evaluated for its effects on bronchial epithelial cells (Calu-3 cells). It was demonstrated that the effect of TJMs especially Na decanoate resulted in a reversible opening of tight junctions evidenced by the decrease in the transepithelial resistance. It was also demonstrated that this reduction of TEER upon exposing the epithelial cells to the TJMs resulted in a significant increase in Flu-Na (paracellular marker) and PXS25 (anti-fibrotic compound) transepithelial transport through this barrier. In conclusion, among the investigated non-specific TJMs, Na decanoate fulfilled the requirements of an effective, non-toxic and reversible tight junction modulator for Calu-3 lung epithelial cells.


Assuntos
Brônquios/metabolismo , Permeabilidade da Membrana Celular/efeitos dos fármacos , Junções Íntimas/efeitos dos fármacos , Transporte Biológico , Biomarcadores/metabolismo , Brônquios/citologia , Linhagem Celular , Células Epiteliais/metabolismo , Humanos , Manosefosfatos/metabolismo
14.
PLoS One ; 11(2): e0149391, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26890638

RESUMO

Among the human heme-peroxidase family, myeloperoxidase (MPO) has a unique disulfide-linked oligomeric structure resulting from multi-step processing of the pro-protein monomer (proMPO) after it exits the endoplasmic reticulum (ER). Related family members undergo some, but not all, of the processing steps involved with formation of mature MPO. Lactoperoxidase has its pro-domain proteolytically removed and is a monomer in its mature form. Eosinophil peroxidase undergoes proteolytic removal of its pro-domain followed by proteolytic separation into heavy and light chains and is a heterodimer. However, only MPO undergoes both these proteolytic modifications and then is further oligomerized into a heterotetramer by a single inter-molecular disulfide bond. The details of how and where the post-ER processing steps of MPO occur are incompletely understood. We report here that T47D breast cancer cells stably transfected with an MPO expression plasmid are able to efficiently replicate all of the processing steps that lead to formation of the mature MPO heterotetramer. MPO also traffics to the lysosome granules of T47D cells where it accumulates, allowing in-depth immunofluorescent microscopy studies of MPO trafficking and storage for the first time. Using this novel cell model we show that formation of MPO's single inter-molecular disulfide bond can occur normally in the absence of the proteolytic events that lead to separation of the MPO heavy and light chains. We further demonstrate that Cys319, which forms MPO's unique inter-molecular disulfide bond, is important for events that precede this step. Mutation of this residue alters the glycosylation and catalytic activity of MPO and blocks its entry into the endocytic pathway where proteolytic processing and disulfide bonding occur. Finally, using the endocytic trafficking of lysosomal hydrolases as a guide, we investigate the role of candidate receptors in the endocytic trafficking of MPO.


Assuntos
Expressão Gênica , Lisossomos/metabolismo , Peroxidase/genética , Peroxidase/metabolismo , Processamento de Proteína Pós-Traducional , Catepsinas/antagonistas & inibidores , Linhagem Celular , Linhagem Celular Tumoral , Cistina/metabolismo , Dissulfetos , Retículo Endoplasmático , Ativação Enzimática , Técnicas de Silenciamento de Genes , Glicosilação , Humanos , Espaço Intracelular , Manosefosfatos/metabolismo , Mutação , Peroxidase/química , Multimerização Proteica , Transporte Proteico , Proteólise , RNA Interferente Pequeno/genética
15.
J Inherit Metab Dis ; 39(2): 293-303, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26310963

RESUMO

Enzyme replacement therapy (ERT) is an effective treatment for several lysosomal storage disorders (LSDs). Intravenously infused enzymes are taken up by tissues through either the mannose 6-phosphate receptor (M6PR) or the mannose receptor (MR). It is generally believed that M6PR-mediated endocytosis is a key mechanism for ERT in treating LSDs that affect the non-macrophage cells of visceral organs. However, the therapeutic efficacy of MR-mediated delivery of mannose-terminated enzymes in these diseases has not been fully evaluated. We tested the effectiveness of a non-phosphorylated α-galactosidase A produced from moss (referred to as moss-aGal) in vitro and in a mouse model of Fabry disease. Endocytosis of moss-aGal was MR-dependent. Compared to agalsidase alfa, a phosphorylated form of α-galactosidase A, moss-aGal was more preferentially targeted to the kidney. Cellular localization of moss-aGal and agalsidase alfa in the heart and kidney was essentially identical. A single injection of moss-aGal led to clearance of accumulated substrate in the heart and kidney to an extent comparable to that achieved by agalsidase alfa. This study suggested that mannose-terminated enzymes may be sufficiently effective for some LSDs in which non-macrophage cells are affected, and that M6P residues may not always be a prerequisite for ERT as previously considered.


Assuntos
Doença de Fabry/enzimologia , Doença de Fabry/metabolismo , Lectinas Tipo C/metabolismo , Lectinas de Ligação a Manose/metabolismo , Manosefosfatos/metabolismo , Receptores de Superfície Celular/metabolismo , alfa-Galactosidase/metabolismo , Animais , Linhagem Celular , Modelos Animais de Doenças , Terapia de Reposição de Enzimas/métodos , Feminino , Humanos , Isoenzimas/metabolismo , Rim/metabolismo , Doenças por Armazenamento dos Lisossomos/enzimologia , Doenças por Armazenamento dos Lisossomos/metabolismo , Masculino , Receptor de Manose , Camundongos , Camundongos Endogâmicos C57BL , Receptor IGF Tipo 2/metabolismo , Proteínas Recombinantes
16.
Traffic ; 16(7): 743-59, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25786328

RESUMO

Most lysosomal enzymes require mannose 6-phosphate (M6P) residues for efficient receptor-mediated lysosomal targeting. Although the lack of M6P residues results in missorting and hypersecretion, selected lysosomal enzymes reach normal levels in lysosomes of various cell types, suggesting the existence of M6P-independent transport routes. Here, we quantify the lysosomal proteome in M6P-deficient mouse fibroblasts (PT(ki)) using Stable Isotope Labeling by Amino acids in Cell culture (SILAC)-based comparative mass spectrometry, and find unchanged amounts of 20% of lysosomal enzymes, including cathepsins D and B (Ctsd and Ctsb). Examination of fibroblasts from a new mouse line lacking both M6P and sortilin, a candidate for M6P-independent transport of lysosomal enzymes, revealed that sortilin does not act as cargo receptor for Ctsb and Ctsd. Using fibroblast lines deficient for endocytic lipoprotein receptors, we could demonstrate that both LDL receptor and Lrp1 mediate the internalization of non-phosphorylated Ctsb and Ctsd. Furthermore, the presence of Lrp1 inhibitor increased the secretion of Ctsd from PT(ki) cells. These findings establish Lrp1 and LDL receptors in M6P-independent secretion-recapture targeting mechanism for lysosomal enzymes.


Assuntos
Lisossomos/metabolismo , Manosefosfatos/metabolismo , Receptores de LDL/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Catepsinas/metabolismo , Células Cultivadas , Fibroblastos/metabolismo , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Camundongos , Camundongos Endogâmicos C57BL , Transporte Proteico , Receptores de LDL/antagonistas & inibidores , Receptores de LDL/genética , Proteínas Supressoras de Tumor/antagonistas & inibidores , Proteínas Supressoras de Tumor/genética
17.
Glycobiology ; 25(6): 591-606, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25573276

RESUMO

The 300 kDa cation-independent mannose 6-phosphate receptor (CI-MPR) plays an essential role in lysosome biogenesis by targeting ∼ 60 different phosphomannosyl-containing acid hydrolases to the lysosome. This type I membrane glycoprotein has a large extracellular region comprised of 15 homologous domains. Two mannose 6-phosphate (M6P) binding sites have been mapped to domains 3 and 9, whereas domain 5 binds preferentially to the phosphodiester, M6P-N-acetylglucosamine (GlcNAc). A structure-based sequence alignment predicts that the C-terminal domain 15 contains three out of the four conserved residues identified as essential for carbohydrate recognition by domains 3, 5 and 9 of the CI-MPR, but lacks two cysteine residues that are predicted to form a disulfide bond. To determine whether domain 15 of the CI-MPR has lectin activity and to probe its carbohydrate-binding specificity, truncated forms of the CI-MPR were tested for binding to acid hydrolases with defined N-glycans in surface plasmon resonance analyses, and used to interrogate a phosphorylated glycan microarray. The results show that a construct encoding domains 14-15 binds both M6P and M6P-GlcNAc with similar affinity (Kd = 13 and 17 µM, respectively). Site-directed mutagenesis studies demonstrate the essential role of the conserved Tyr residue in domain 15 for phosphomannosyl binding. A structural model of domain 15 was generated that predicted an Arg residue to be in the binding pocket and mutagenesis studies confirmed its important role in carbohydrate binding. Together, these results show that the CI-MPR contains a fourth carbohydrate-recognition site capable of binding both phosphomonoesters and phosphodiesters.


Assuntos
Manosefosfatos/metabolismo , Receptor IGF Tipo 2/química , Receptor IGF Tipo 2/metabolismo , Animais , Sítios de Ligação , Cátions , Bovinos , Hidrolases/metabolismo , Análise em Microsséries , Modelos Moleculares , Ressonância de Plasmônio de Superfície
18.
Mol Genet Metab ; 114(2): 268-73, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25468650

RESUMO

Fabry disease (FD) is an X-linked lysosomal storage disorder resulting from deficiency of α-galactosidase A (GLA). Traditionally, heterozygotes were considered asymptomatic carriers of FD, but it is now apparent that the asymptomatic female carrier is the exception and most heterozygotes suffer significant multisystemic disease. To determine why the process of cross-correction does not occur effectively in FD heterozygotes, we investigated GLA production and secretion in cultured skin fibroblasts as well as GLA levels in plasma. The maturation of GLA was similar in FD heterozygotes and control fibroblasts, confirming that both produce the 46kDa mature form; the same as that present in control plasma. However, the proportion of GLA secreted into the culture media was substantially less than eight other lysosomal proteins. Artificial generation of FD heterozygotes in cellulo, along with another lysosomal storage disorder, mucopolysaccharidosis type II, revealed no cross-correction in the FD system, whereas MPS II fibroblasts were able to cross-correct. In plasma, GLA was present as the 46kDa mature form, which lacks the mannose 6-phosphorylated moiety and is not able to be efficiently endocytosed by affected cells. Our evidence shows that fibroblasts secrete minimal amounts of GLA and consequently normal fibroblasts are unable to cross-correct FD fibroblasts. We suggest that symptomatic FD heterozygotes arise due to the secretion of primarily the mature form, with only small amounts of the mannose 6-phosphorylated form of GLA from unaffected cells. This limits capacity for enzyme cross correction of affected cells, despite uptake of exogenous recombinant GLA.


Assuntos
Doença de Fabry/enzimologia , Heterozigoto , alfa-Galactosidase/metabolismo , Células Cultivadas , Doença de Fabry/genética , Fibroblastos/enzimologia , Humanos , Manosefosfatos/metabolismo , Mucopolissacaridose II/enzimologia , Pele/citologia , Pele/enzimologia , Triexosilceramidas/metabolismo , alfa-Galactosidase/sangue
19.
J Biol Chem ; 289(18): 12232-44, 2014 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-24573685

RESUMO

The overproduction of polysaccharide alginate is responsible for the formation of mucus in the lungs of cystic fibrosis patients. Histidine kinase KinB of the KinB-AlgB two-component system in Pseudomonas aeruginosa acts as a negative regulator of alginate biosynthesis. The modular architecture of KinB is similar to other histidine kinases. However, its periplasmic signal sensor domain is unique and is found only in the Pseudomonas genus. Here, we present the first crystal structures of the KinB sensor domain. The domain is a dimer in solution, and in the crystal it shows an atypical dimer of a helix-swapped four-helix bundle. A positively charged cavity is formed on the dimer interface and involves several strictly conserved residues, including Arg-60. A phosphate anion is bound asymmetrically in one of the structures. In silico docking identified several monophosphorylated sugars, including ß-D-fructose 6-phosphate and ß-D-mannose 6-phosphate, a precursor and an intermediate of alginate synthesis, respectively, as potential KinB ligands. Ligand binding was confirmed experimentally. Conformational transition from a symmetric to an asymmetric structure and decreasing dimer stability caused by ligand binding may be a part of the signal transduction mechanism of the KinB-AlgB two-component system.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas Quinases/metabolismo , Pseudomonas aeruginosa/metabolismo , Fatores de Transcrição/metabolismo , Alginatos/química , Sequência de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Sítios de Ligação/genética , Dicroísmo Circular , Cristalografia por Raios X , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Frutosefosfatos/química , Frutosefosfatos/metabolismo , Ácido Glucurônico/biossíntese , Ácido Glucurônico/química , Ácidos Hexurônicos/química , Histidina Quinase , Ligantes , Manosefosfatos/química , Manosefosfatos/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Proteínas Quinases/química , Proteínas Quinases/genética , Multimerização Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Pseudomonas aeruginosa/genética , Homologia de Sequência de Aminoácidos , Fatores de Transcrição/química , Fatores de Transcrição/genética
20.
Traffic ; 15(5): 500-15, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24502338

RESUMO

The hyaluronidase Hyal-1 is an acid hydrolase that degrades hyaluronic acid (HA), a component of the extracellular matrix. It is often designated as a lysosomal protein. Yet few data are available on its intracellular localization and trafficking. We demonstrate here that in RAW264.7 murine macrophages, Hyal-1 is synthesized as a glycosylated precursor that is only weakly mannose 6-phosphorylated. Nevertheless, this precursor traffics to endosomes, via a mannose 6-phosphate-independent secretion/recapture mechanism that involves the mannose receptor. Once in endosomes, it is processed into a lower molecular mass form that is transported to lysosomes, where its activity could be detected using native gel zymography. Indeed, this activity co-distributed with lysosomal hydrolases in the densest fraction of a self-forming Percoll(TM) density gradient. Moreover, it shifted toward the lower density region, in parallel with those hydrolases, when a decrease of lysosomal density was induced by the endocytosis of sucrose. Interestingly, the activity of the processed form of Hyal-1 was largely underestimated when assayed by zymography after SDS-PAGE and subsequent renaturation of the proteins, by contrast to the full-length protein that could efficiently degrade HA in those conditions. These results suggest that noncovalent associations support the lysosomal activity of Hyal-1.


Assuntos
Hialuronoglucosaminidase/metabolismo , Macrófagos/metabolismo , Transporte Proteico/fisiologia , Animais , Endocitose/fisiologia , Endossomos/metabolismo , Glicosilação , Hidrolases/metabolismo , Lectinas Tipo C/metabolismo , Lisossomos/metabolismo , Receptor de Manose , Lectinas de Ligação a Manose/metabolismo , Manosefosfatos/metabolismo , Camundongos , Receptores de Superfície Celular/metabolismo , Sacarose/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA