Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
1.
Microb Pathog ; 190: 106628, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38508422

RESUMO

Rotavirus infections in suckling and weaning piglets cause severe dehydration and death, resulting in significant economic losses in the pig breeding industry. With the continuous emergence of porcine rotavirus (PoRV) variants and poor vaccine cross-protection among various genotypes, there is an urgent need to develop alternative strategies such as seeking effective antiviral products from nature, microbial metabolites and virus-host protein interaction. Sialidases play a crucial role in various physiopathological processes and offer a promising target for developing antivirus drugs. However, the effect of bacterial-derived sialidases on the infection of PoRVs remains largely unknown. Herein, we investigated the impact of bacterial-derived sialidases (sialidase Cp and Vc) on PoRV strain OSU(Group A) infection, using differentiated epithelial monkey kidney cells (MA104) as a model. Our results indicated that the pretreatment of MA104 with exogenous sialidases effectively suppressed PoRV OSU in a concentration-dependent manner. Notably, even at a concentration of 0.01 µU/mL, sialidases significantly inhibited the virus (MOI = 0.01). Meanwhile, we found that sialidase Vc pretreatment sharply reduced the binding rate of PoRV OSU. Last, we demonstrated that PoRV OSU might recognize α-2,3-linked sialic acid as the primary attachment factor in MA104. Our findings provide new insights into the underlying mechanism of PoRV OSU infections, shedding lights on the development of alternative antivirus approaches based on bacteria-virus interaction.


Assuntos
Neuraminidase , Infecções por Rotavirus , Rotavirus , Replicação Viral , Animais , Neuraminidase/metabolismo , Neuraminidase/genética , Rotavirus/efeitos dos fármacos , Rotavirus/fisiologia , Suínos , Replicação Viral/efeitos dos fármacos , Linhagem Celular , Células Epiteliais/virologia , Células Epiteliais/microbiologia , Ligação Viral/efeitos dos fármacos , Ácido N-Acetilneuramínico/metabolismo , Ácido N-Acetilneuramínico/farmacologia , Antivirais/farmacologia , Haplorrinos , Doenças dos Suínos/virologia , Doenças dos Suínos/microbiologia
2.
J Immunother Cancer ; 11(11)2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37940346

RESUMO

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers. Despite the successful application of immune checkpoint blockade in a range of human cancers, immunotherapy in PDAC remains unsuccessful. PDAC is characterized by a desmoplastic, hypoxic and highly immunosuppressive tumor microenvironment (TME), where T-cell infiltration is often lacking (immune desert), or where T cells are located distant from the tumor islands (immune excluded). Converting the TME to an immune-inflamed state, allowing T-cell infiltration, could increase the success of immunotherapy in PDAC. METHOD: In this study, we use the KPC3 subcutaneous PDAC mouse model to investigate the role of tumor-derived sialic acids in shaping the tumor immune landscape. A sialic acid deficient KPC3 line was generated by genetic knock-out of the CMAS (cytidine monophosphate N-acetylneuraminic acid synthetase) enzyme, a critical enzyme in the synthesis of sialic acid-containing glycans. The effect of sialic acid-deficiency on immunotherapy efficacy was assessed by treatment with anti-programmed cell death protein 1 (PD-1) and agonistic CD40. RESULT: The absence of sialic acids in KPC3 tumors resulted in increased numbers of CD4+ and CD8+ T cells in the TME, and reduced frequencies of CD4+ regulatory T cells (Tregs) within the T-cell population. Importantly, CD8+ T cells were able to infiltrate the tumor islands in sialic acid-deficient tumors. These favorable alterations in the immune landscape sensitized sialic acid-deficient tumors to immunotherapy, which was ineffective in sialic acid-expressing KPC3 tumors. In addition, high expression of sialylation-related genes in human pancreatic cancer correlated with decreased CD8+ T-cell infiltration, increased presence of Tregs, and poorer survival probability. CONCLUSION: Our results demonstrate that tumor-derived sialic acids mediate T-cell exclusion within the PDAC TME, thereby impairing immunotherapy efficacy. Targeting sialic acids represents a potential strategy to enhance T-cell infiltration and improve immunotherapy outcomes in PDAC.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Camundongos , Animais , Humanos , Linfócitos T CD8-Positivos , Ácidos Siálicos/farmacologia , Ácido N-Acetilneuramínico/farmacologia , Neoplasias Pancreáticas/metabolismo , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/genética , Imunoterapia/métodos , Microambiente Tumoral
3.
Bioconjug Chem ; 34(8): 1498-1507, 2023 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-37498932

RESUMO

Cell communication and signal transduction rely heavily on the charge on the cell surface. The cell surface is negatively charged, with glycoproteins on the cell membrane providing a large percentage of the charge. Sialic acid is found on the outermost side of glycan chains and contributes to glycoprotein's negative charge. Sialic acid is highly expressed in tumor cells and plays an important role in tumor metastasis and immune escape by interacting with extracellular ligands. However, the specific effect of negative charge changes on glycoproteins is still poorly understood. In this study, we used 9-azido sialic acid (9Az-Sia) to create artificial epitopes on glycoproteins via metabolic glycan labeling, and we attached charged groups such as amino and carboxyl to 9Az-Sia via a click reaction with dibenzocyclooctyne (DBCO). The charge of glycoproteins was changed by metabolic glycan labeling and click modification. The results showed that the migration and invasion ability of the MDA-MB-231 cell labeled with 9Az-Sia was significantly reduced after the modification with amino groups rather than carboxyl groups. Epithelial-mesenchymal transition (EMT) is the biological process of metastatic tumor cells, with an increasing ability of tumor cells to migrate and invade. In particular, the expression of adhesion molecules increased in the amine-linked group, whereas the expression of matrix metalloproteinases (MMPs) increased significantly, which is not identical to EMT characteristics. In vivo experiments have demonstrated that the loss of negative charge on glycoproteins has an inhibitory effect on tumors. In conclusion, modifying the positive charge on the surface of glycoproteins can inhibit tumor cell metastasis and has great potential for tumor therapy.


Assuntos
Glicoproteínas de Membrana , Neoplasias , Humanos , Ácido N-Acetilneuramínico/farmacologia , Transição Epitelial-Mesenquimal , Movimento Celular , Neoplasias/patologia , Glicoproteínas , Metaloproteinases da Matriz , Junções Intercelulares/patologia , Linhagem Celular Tumoral , Metástase Neoplásica/patologia
4.
Eur J Pharm Biopharm ; 184: 50-61, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36682511

RESUMO

Although anti-tumor strategies targeting tumor-associated immune cells were being rapidly developed, the preparations were usually limited in targeting efficiency. To overcome this barrier, this study reported a novel sialic acid-octadecylamine (SA-ODA) and monosialotetrahexosylganglioside (GM1) co-modified epirubicin liposomes (5-5-SAGL-EPI), which improved tumor-targeting ability through the active targeting of tumor-associated macrophages (TAMs) by SA-ODA and the long circulation of GM1. Thus, we evaluated 5-5-SAGL-EPI in vitro and in vivo. Analysis of cellular uptake by RAW264.7 cells using flow cytometry and confocal microscopy showed a higher rate of cellular uptake for 5-5-SAGL-EPI than for the common liposomes (CL-EPI). In pharmacokinetic studies using Wistar rats, compared to CL-EPI, 5-5-SAGL-EPI showed a higher circulation time in vivo. Tissue distribution studies in Kunming mice bearing S180 tumors revealed increased distribution of 5-5-SAGL-EPI in tumor tissues compared with liposomes modified with single ligands (SA-ODA [5-SAL-EPI] or GM1 [5-GL-EPI]). In vivo anti-tumor experiments using the S180 tumor-bearing mice revealed a high tumor inhibition rate and low toxicity for 5-5-SAGL-EPI. Moreover, freeze-dried 5-5-SAGL-EPI had good storage stability, and the anti-tumor effect was comparable to that before freeze-drying. Overall, 5-5-SAGL-EPI exhibited excellent anti-tumor effects before and after lyophilization.


Assuntos
Lipossomos , Ácido N-Acetilneuramínico , Camundongos , Ratos , Animais , Lipossomos/farmacologia , Ácido N-Acetilneuramínico/farmacologia , Macrófagos Associados a Tumor , Microambiente Tumoral , Gangliosídeo G(M1)/farmacologia , Ratos Wistar , Linhagem Celular Tumoral
5.
Pathol Res Pract ; 240: 154186, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36327814

RESUMO

The aim of current work was able to show the oxidant effect of cancer cells found in any part of the body on the liver and to investigate the possible protective effect of deuterium-depleted water (DDW) on this oxidant effect by determining of some liver parameters. Ehrlich ascites tumor bearing BALB/c mice were used for this purpose. BALB/c mice were selected randomly and divided into four groups (n = 5 in each group) as control group, tumor group, control+DDW group, tumor+DDW group, fifteen days after tumor cell injection, liver tissue samples were taken for all groups. In the tumor group, liver lipid peroxidation, sialic acid and protein carbonyl levels, xanthine oxidase, myeloperoxidase, catalase, gamma-glutamyl transferase, sorbitol dehydrogenase, glutathione peroxidase and glutathione reductase activities, were significantly higher than those in the control group while glutathione levels and paraoxonase1, sodium potassium ATPase, glutathione-S-transferase, alanine transaminase and aspartate transaminase activities decreased significantly. Compared with the tumor group, the changes in all parameters except sialic acid, catalase, alanine transaminase and aspartate transaminase were reversed in the DDW given tumor groups, while sialic acid and catalase values continued to increase, and alanine transaminase and aspartate transaminase values continued to decrease. In conclusion, the consumption of DDW may be beneficial and protective against excessive oxidative stress in cancer complications.


Assuntos
Água Potável , Camundongos , Animais , Catalase/metabolismo , Alanina Transaminase/metabolismo , Alanina Transaminase/farmacologia , Água Potável/metabolismo , Deutério/metabolismo , Deutério/farmacologia , Ácido N-Acetilneuramínico/metabolismo , Ácido N-Acetilneuramínico/farmacologia , Estresse Oxidativo , Aspartato Aminotransferases/metabolismo , Aspartato Aminotransferases/farmacologia , Peroxidação de Lipídeos , Antioxidantes/farmacologia , Glutationa/metabolismo , Fígado/patologia , Glutationa Transferase , Oxidantes/metabolismo , Oxidantes/farmacologia , Superóxido Dismutase/metabolismo
6.
Life Sci ; 310: 121081, 2022 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-36273630

RESUMO

AIMS: Sialic acid derivatives (SA-derivatives) provide a nanomedicine platform for tumor-targeted delivery and treatment, and allow modulation of immunosuppressive tumor microenvironments with excellent therapeutic effects. Further, the multi-reactive groups of sialic acid (SA) contribute to the diversity of SA derivatives, which inevitably has implications for drug delivery systems and tumor therapy. However, relevant research remains lacking at present. Therefore, this study aimed to explore the effects of SA derivatives on SA-mediated drug delivery systems. MAIN METHODS: Four SA-derivatives with different linking bonds (ester and amide bonds), different linking groups (hydroxyl and carboxyl), and different linking objects (cholesterol, octadecanoic acid, and octadecylamine) were synthesized and the respective SA derivative-modified doxorubicin liposomes were prepared. In-depth research was conducted using both cells and animals. KEY FINDINGS: We found that an SA-cholesterol conjugate (SA-CH; linking bond, amide bond; linking group, carboxyl; linking object, cholesterol) could improve liposome stability, reduce liposome adsorption to plasma proteins, and enhance the targeting of liposomes for killing tumor-associated macrophages (TAMs). Reduced TAMs in the immunosuppressive tumor microenvironment lead to enhanced tumor infiltration of CD8+ T cells. SIGNIFICANCE: The results of this experiment provide clarity for research and development on SA-derivatives and a theoretical basis for clinical trials of SA-derivative-modified nanoparticles.


Assuntos
Lipossomos , Neoplasias , Animais , Lipossomos/química , Microambiente Tumoral , Ácido N-Acetilneuramínico/química , Ácido N-Acetilneuramínico/farmacologia , Linfócitos T CD8-Positivos/metabolismo , Neoplasias/tratamento farmacológico , Colesterol/química , Amidas/farmacologia , Linhagem Celular Tumoral
7.
Yakugaku Zasshi ; 142(10): 1045-1053, 2022.
Artigo em Japonês | MEDLINE | ID: mdl-36184438

RESUMO

Sialic acid-binding lectin from Rana catesbeiana (cSBL) is a multifunctional protein with both lectin and ribonuclease activity and is, therefore, called a leczyme. It exerts cancer cell-selective antitumor effects on a variety of cancer cells in vitro and in vivo under conditions where no undesired side effects are observed. cSBL elicits antitumor effects by degrading cellular RNA and subsequently inducing apoptosis via a pathway mediated by mitochondria and endoplasmic reticulum stress. Further, it exerts synergistic antitumor effects with other molecules such as tumor necrosis factor-related apoptosis-inducing ligand and pemetrexed. Recent studies have revealed that long-term treatment of cancer cells with cSBL causes significant pleiotropic changes in the expression profiles of several genes, including multiple genes involved in metabolic pathways. Furthermore, cSBL reduces the expression of some cancer-related molecules such as human epidermal growth factor receptors, aldo-keto reductase 1B10, and ATP-binding cassette transporter C2. The information described above is expected to lead to useful applications, such as effective regimens comprising cSBL and other drugs. These findings reveal favorable properties of cSBL as an anticancer drug, which may contribute to the development of new therapeutic strategies for cancer treatment.


Assuntos
Antineoplásicos , Neoplasias , Transportadores de Cassetes de Ligação de ATP , Aldo-Ceto Redutases/metabolismo , Animais , Antineoplásicos/uso terapêutico , Apoptose , Linhagem Celular Tumoral , Família de Proteínas EGF/metabolismo , Família de Proteínas EGF/farmacologia , Humanos , Lectinas/química , Ligantes , Ácido N-Acetilneuramínico/farmacologia , Neoplasias/tratamento farmacológico , Pemetrexede , RNA/farmacologia , Rana catesbeiana/metabolismo , Ribonucleases/metabolismo , Fatores de Necrose Tumoral/metabolismo , Fatores de Necrose Tumoral/farmacologia
8.
J Biochem Mol Toxicol ; 36(9): e23124, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35670011

RESUMO

Galactosamine (GalN) is a well-known agent for inducing viral hepatitis models in rodents, but it can cause toxicity on different organs. Vitamin U (Vit U) has been proved as a powerful antioxidant on many toxicity models. The present study was designed to investigate the protective effects of Vit U on GalN-induced stomach injury. Rats were divided into four groups as follows: control (group I), Vit U given animals (50 mg/kg per day; group II), GalN administered animals (500 mg/kg at a single dose; group III), GalN + Vit U given animals (at the same dose and time, group IV). At the end of the 3rd day, animals were killed, and stomach tissues were taken. They were homogenized and centrifuged. In comparison to the control group, glutathione, total antioxidant capacity levels, catalase, superoxide dismutase, glutathione peroxidase, glutathione reductase, glutathione-S-transferase, and Na+ /K+ -ATPase activities of GalN group were found to be decreased. On the contrary, lipid peroxidation, advanced oxidized protein products, hexose-hexosamine, fucose, sialic acid, reactive oxygen species levels, as well as the activities of myeloperoxidase, xanthine oxidase, and lactate dehydrogenase were elevated. Administration of Vit U reversed these abnormalities in the GalN group. It can be concluded that Vit U exerts its unique antioxidant effect and prevents GalN-induced gastric damage.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas , Vitamina U , Adenosina Trifosfatases/metabolismo , Adenosina Trifosfatases/farmacologia , Animais , Antioxidantes/metabolismo , Antioxidantes/farmacologia , Catalase/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Fucose/farmacologia , Galactosamina/toxicidade , Glutationa/metabolismo , Glutationa Peroxidase/metabolismo , Glutationa Redutase/metabolismo , Glutationa Transferase/metabolismo , Lactato Desidrogenases/metabolismo , Peroxidação de Lipídeos , Ácido N-Acetilneuramínico/farmacologia , Estresse Oxidativo , Peroxidase/metabolismo , Ratos , Espécies Reativas de Oxigênio , Superóxido Dismutase/metabolismo , Vitamina U/farmacologia , Xantina Oxidase/metabolismo
9.
Biol Bull ; 241(2): 196-207, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34706210

RESUMO

AbstractSensory receptors control nematocyst discharge on sea anemone tentacles. Micromolar N-acetylated sugars (e.g., N-acetyl neuraminic acid [NANA]) bind chemoreceptors on ectodermal supporting cells and predispose adjacent nematocyst discharge in response to mechanical contact via a cyclic adenosine monophosphate (cAMP)-dependent sensitization pathway, while higher NANA levels dose-dependently desensitize. Recent evidence implicates L-type calcium channels in desensitizing the pathway in aconitate sea anemones Aiptasia pallida (also known as Exaiptasia diaphana). We, therefore, hypothesize that NANA activates calcium influx via L-type calcium channels. We demonstrate a dose-dependent, NANA-activated 45Ca influx into dissociated ectodermal cells isolated from A. pallida tentacles, with maximal influx occurring at desensitizing concentrations of NANA. The L-type calcium channel inhibitors nifedipine, diltiazem, methoxyverapamil, and cadmium blocked NANA-stimulated 45Ca influx. Elevated extracellular KCl levels dose-dependently increased nifedipine-sensitive 45Ca influx to implicate voltage-gated calcium channels. Forskolin, 8-bromo-cAMP, and the protein kinase A inhibitor H-8 affect NANA-stimulated calcium influx in a manner consistent with activated cAMP-dependent pathway involvement. Because NANA chemoreceptors localize to supporting cells of cnidocyte supporting cell complexes, NANA activation of 45Ca influx into isolated tentacle ectodermal cells suggests that L-type calcium channels and NANA chemoreceptors co-localize to supporting cells. Indeed, a fluorescent marker of L-type calcium channels localizes to the apical ectoderm adjacent to nematocysts of live tentacles. We conclude that supporting cell chemoreceptors activate co-localized L-type calcium channels via a cAMP-dependent mechanism in order to initiate desensitization. We suggest that pathway desensitization may conserve nematocysts from excessive discharge during prey capture.


Assuntos
Canais de Cálcio Tipo L/efeitos dos fármacos , Ácido N-Acetilneuramínico/farmacologia , Anêmonas-do-Mar , Animais , Cálcio , AMP Cíclico , Nematocisto
10.
Drug Deliv ; 28(1): 1849-1860, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34515617

RESUMO

Melanoma is one of the most common malignant tumors. The anti-PD-1 antibody is used for the treatment of metastatic melanoma. Treatment success is only 35-40% and a range of immune-related adverse reactions can occur. Combination of anti-PD1 antibody therapy with other oncology therapies has been attempted. Herein, we assessed whether chlorogenic acid liposomes modified with sialic acid (CA-SAL) combined with anti-PD1 antibody treatment was efficacious as immunotherapy for melanoma. CA-SAL liposomes were prepared and characterized. In a mouse model of B16F10 tumor, mice were treated with an anti-PD1 antibody, CA-SAL, or combination of CA-SAL + anti-PD1 antibody, and compared with no treatment controls. The tumor inhibition rate, tumor-associated macrophages (TAMs) phenotype, T-cell activity, and safety were investigated. We observed a significant decrease in the proportion of M2-TAMs and CD4+Fop3+ T cells, while there was a significant increase in the proportion of M1-TAMs and CD8+ T cells, and in the activity of T cells, and thus in the tumor inhibition rate. No significant toxicity was observed in major organs. CA-SAL and anti-PD1 Ab combination therapy presented synergistic anti-tumor activity, which enhanced the efficacy of the PD-1 checkpoint blocker in a mouse model of melanoma. In summary, combination immunotherapy of CA-SAL and anti-PD1 Ab has broad prospects in improving the therapeutic effect of melanoma, and may provide a new strategy for clinical treatment.


Assuntos
Ácido Clorogênico/farmacologia , Inibidores de Checkpoint Imunológico/farmacologia , Lipossomos/química , Melanoma/patologia , Ácido N-Acetilneuramínico/farmacologia , Animais , Sobrevivência Celular , Ácido Clorogênico/administração & dosagem , Ácido Clorogênico/farmacocinética , Portadores de Fármacos/química , Combinação de Medicamentos , Inibidores de Checkpoint Imunológico/administração & dosagem , Inibidores de Checkpoint Imunológico/farmacocinética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ácido N-Acetilneuramínico/administração & dosagem , Ácido N-Acetilneuramínico/farmacocinética , Fenótipo , Células RAW 264.7 , Linfócitos T/efeitos dos fármacos , Macrófagos Associados a Tumor/efeitos dos fármacos
11.
Mol Med Rep ; 23(6)2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33880588

RESUMO

Malignant pleural mesothelioma (MPM) is a universally lethal type of cancer that is increasing in incidence worldwide; therefore, the development of new drugs for MPM is an urgent task. Bullfrog sialic acid­binding lectin (cSBL) is a multifunctional protein that has carbohydrate­binding and ribonuclease activities. cSBL exerts marked antitumor activity against numerous types of cancer cells, with low toxicity to normal cells. Although in vitro and in vivo studies revealed that cSBL was effective against MPM, the mechanism by which cSBL exerts antitumor effects is not fully understood. To further understand the mechanism of action of cSBL, the present study aimed to identify the key molecules whose expression was affected by cSBL. The present study established cSBL­resistant MPM cells. Microarray analyses revealed that there were significant pleiotropic changes in the expression profiles of several genes, including multiple genes involved in metabolic pathways in cSBL­resistant cells. Furthermore, the expression of some members of the aldo­keto reductase family was revealed to be markedly downregulated in these cells. Among these, it was particularly interesting that cSBL action reduced the level of AKR1B10, which has been reported as a biomarker candidate for MPM prognosis. These findings revealed novel aspects of the effect of cSBL, which may contribute to the development of new therapeutic strategies for MPM.


Assuntos
Lectinas/farmacologia , Mesotelioma Maligno/tratamento farmacológico , Mesotelioma Maligno/genética , Mesotelioma Maligno/metabolismo , Ácido N-Acetilneuramínico/farmacologia , Rana catesbeiana/metabolismo , Transcriptoma , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Mesotelioma/metabolismo , Prognóstico
12.
Adv Drug Deliv Rev ; 171: 62-76, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33548302

RESUMO

Biological drugs, specifically proteins and peptides, are a privileged class of medicinal agents and are characterized with high specificity and high potency of therapeutic activity. However, biologics are fragile and require special care during storage, and are often modified to optimize their pharmacokinetics in terms of proteolytic stability and blood residence half-life. In this review, we showcase glycosylation as a method to optimize biologics for storage and application. Specifically, we focus on chemical glycosylation as an approach to modify biological drugs. We present case studies that illustrate the success of this methodology and specifically address the highly important question: does connectivity within the glycoconjugate have to be native or not? We then present the innovative methods of chemical glycosylation of biologics and specifically highlight the emerging and established protecting group-free methodologies of glycosylation. We discuss thermodynamic origins of protein stabilization via glycosylation, and analyze in detail stabilization in terms of proteolytic stability, aggregation upon storage and/or heat treatment. Finally, we present a case study of protein modification using sialic acid-containing glycans to avoid hepatic clearance of biological drugs. This review aims to spur interest in chemical glycosylation as a facile, powerful tool to optimize proteins and peptides as medicinal agents.


Assuntos
Produtos Biológicos/química , Animais , Produtos Biológicos/farmacologia , Eritropoetina/química , Eritropoetina/farmacologia , Glicosilação , Humanos , Polipeptídeo Amiloide das Ilhotas Pancreáticas/química , Polipeptídeo Amiloide das Ilhotas Pancreáticas/farmacologia , Ácido N-Acetilneuramínico/química , Ácido N-Acetilneuramínico/farmacologia , Polissacarídeos/química , Polissacarídeos/farmacologia
13.
Am J Physiol Renal Physiol ; 320(4): F559-F568, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33615893

RESUMO

Hypercalciuria is one of the early manifestations of diabetic nephropathy (DN). This is partially due to a decrease in the expression of renal transient receptor potential vanilloid type 5 (TRPV5), which is responsible for renal Ca2+ reabsorption. Soluble klotho has been previously determined to increase TRPV5 by cleaving sialic acid, causing TRPV5 to bind to membrane protein galectin-1. However, a recent study showed that soluble klotho binds to α2-3-sialyllactose, where sialic acid is located, on TRPV5, rather than cleave it. Here, we report that soluble klotho tethers TRPV5 on the membrane by binding both TRPV5 and galectin-1, thereby protecting membrane TRPV5 from diabetes-induced endocytosis. In the present study, we injected recombinant soluble α-klotho protein (rKL) into db/db and db/m mice for 8 wk and collected urine and kidneys. We administered rKL, AZD4547 [fibroblast growth factor (FGF) receptor type 1 inhibitor], and OTX008 (galectin-1 inhibitor) to cultured mouse distal tubular cells with or without 30 mM high-glucose (HG) exposure. db/db mice showed increased renal Ca2+ excretion and decreased renal TRPV5 expression. rKL treatment reversed this change. In vitro, TRPV5 expression in distal tubular cells decreased under HG conditions, and rKL successfully upregulated TRPV5 with or without FGF23. Also, immunofluorescence showed colocalization of klotho, TRPV5, and galectin-1 in distal tubule cells, suggesting that klotho binds to both TRPV5 and galectin-1. Moreover, when both FGF receptor type 1 and galectin-1 were inhibited, rKL failed to increase TRPV5 under HG conditions. Our results indicate that soluble klotho prevents TRPV5 from degradation and subsequent diabetes-induced endocytosis by anchoring TRPV5 through binding with both TRPV5 and galectin-1.NEW & NOTEWORTHY Soluble α-klotho anchors transient receptor potential vanilloid type 5 (TRPV5) on the apical membrane of the distal tubule by binding both TRPV5 and a membrane-abundant protein, galectin-1. This newly discovered mechanism works even when fibroblast growth factor (FGF)23 signaling is inhibited by treatment with FGF receptor type 1 inhibitor. Therefore, we identified how soluble α-klotho increases TRPV5 without FGF23. We confirmed this mechanism by observing that soluble α-klotho fails to enhance TRPV5 when both FGF receptor type 1 and galectin-1 are inhibited.


Assuntos
Canais de Cálcio/efeitos dos fármacos , Canais de Cálcio/metabolismo , Membrana Celular/efeitos dos fármacos , Galectina 1/metabolismo , Rim/metabolismo , Canais de Cátion TRPV/efeitos dos fármacos , Canais de Cátion TRPV/metabolismo , Animais , Benzamidas/farmacologia , Membrana Celular/metabolismo , Nefropatias Diabéticas/metabolismo , Endocitose/efeitos dos fármacos , Endocitose/fisiologia , Células Epiteliais/metabolismo , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/metabolismo , Galectina 1/farmacologia , Camundongos , Ácido N-Acetilneuramínico/farmacologia , Piperazinas/farmacologia , Pirazóis/farmacologia
14.
Lett Appl Microbiol ; 73(1): 20-25, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33386625

RESUMO

Sialic acid (N-acetylneuraminic acid), a 9-carbon monosaccharide, has been widely studied in immunology, oncology and neurology. However, the effects of sialic acid on organ and intestinal development, liver function and gut microbiota were rarely studied. In this study, we found that oral sialic acid tended to increase the relative weight of liver and decreased the serum aspartate aminotransferase (GPT) activity. In addition, sialic acid treatment markedly reduced gut villus length, depth, the ratio of villus length/depth (L/D), areas, width and the number of goblet cells. Furthermore, gut microbes were changed in response to oral sialic acid, such as Staphylococcus lentus, Corynebacterium stationis, Corynebacterium urealyticum, Jeotgalibaca sp_PTS2502, Ignatzschineria indica, Sporosarcina pasteurii, Sporosarcina sp_HW10C2, Facklamia tabacinasalis, Oblitimonas alkaliphila, Erysipelatoclostridium ramosum, Blautia sp_YL58, Bacteroids thetaiotaomicron, Morganella morganii, Clostridioides difficile, Helicobacter tryphlonius, Clostridium sp_Clone47, Alistipes finegoldii, [pseudomonas]_geniculata and Pseudomonas parafulva at the species level. In conclusion, oral sialic acid altered the intestinal pathological state and microbial compositions, and the effect of sialic acid on host health should be further studied.


Assuntos
Biodiversidade , Microbioma Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/efeitos dos fármacos , Fígado/efeitos dos fármacos , Ácido N-Acetilneuramínico/farmacologia , Administração Oral , Animais , Aspartato Aminotransferases/sangue , Ativação Enzimática/efeitos dos fármacos , Trato Gastrointestinal/crescimento & desenvolvimento , Camundongos , Ácido N-Acetilneuramínico/administração & dosagem
15.
PLoS One ; 15(12): e0244762, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33378413

RESUMO

Non-alcoholic fatty liver disease (NAFLD) is associated with obesity and type 2 diabetes and is characterized by the accumulation of fat in the liver (steatosis). NAFLD can transition into non-alcoholic steatohepatitis (NASH), with liver cell injury, inflammation, and an increased risk of fibrosis. We previously found that injections of either 1866, a synthetic ligand for the lectin receptor CD209, or DANA, a sialidase inhibitor, can inhibit inflammation and fibrosis in multiple animal models. The methionine and choline-deficient (MCD) diet is a model of NASH which results in the rapid induction of liver steatosis and inflammation. In this report, we show that for C57BL/6 mice on a MCD diet, injections of both 1866 and DANA reversed MCD diet-induced decreases in white fat, decreases in adipocyte size, and white fat inflammation. However, these effects were not observed in type 2 diabetic db/db mice on a MCD diet. In db/db mice on a MCD diet, 1866 decreased liver steatosis, but these effects were not observed in C57BL/6 mice. There was no correlation between the ability of 1866 or DANA to affect steatosis and the effects of these compounds on the density of liver macrophage cells expressing CLEC4F, CD64, F4/80, or Mac2. Together these results indicate that 1866 and DANA modulate adipocyte size and adipose tissue macrophage populations, that 1866 could be useful for modulating steatosis, and that changes in the local density of 4 different liver macrophages cell types do not correlate with effects on liver steatosis.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Moléculas de Adesão Celular/agonistas , Lectinas Tipo C/agonistas , Fígado/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Ácido N-Acetilneuramínico/análogos & derivados , Neuraminidase/antagonistas & inibidores , Receptores de Superfície Celular/agonistas , Tecido Adiposo/metabolismo , Animais , Deficiência de Colina/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animais de Doenças , Fígado Gorduroso/tratamento farmacológico , Fígado Gorduroso/metabolismo , Inflamação/metabolismo , Fígado/metabolismo , Macrófagos/metabolismo , Camundongos , Ácido N-Acetilneuramínico/farmacologia , Ácido N-Acetilneuramínico/uso terapêutico , Neuraminidase/metabolismo , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Hepatopatia Gordurosa não Alcoólica/metabolismo
16.
Int Immunopharmacol ; 88: 106881, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32795899

RESUMO

Necrotizing enterocolitis (NEC), a devastating infant disease characterized by severe intestinal necrosis, its pathogenesis is poorly understood, but appears to be multifactorial and highly associated with immaturity of gastrointestinal tract and immature innate-immune system. Breast-milk is effective strategy to protect infants against NEC. This study is using a NEC rat model to investigate the pathological mechanism of NEC involved intestinal-damages, and the therapeutic mechanism of sialylated human milk oligosaccharides (SHMOs) on NEC rats; also using cell model to investigate the effects of SHMOs on colon-epithelial cells (Caco-2) in-vitro. Extraction and characterization of SHMOs from breast milk, establishment of a NEC rat model, histopathological analysis and mast cell accounting of the terminal ileum were taken; The levels of DPPI, TLR4, IL-6, TNF-α, MMP-2/9 and glutathione were measured using various methods. Caco-2 cells were pre-treated with SHMOs and cultured with LPS, histamine, chymase or DPPI, cell viabilities and mitochondrial membrane potential were examined; flow cytometry was used to detect cell cycle. The accumulation of mast cells was found in the ileum of NEC rats, but prohibited by SHMOs treatment; the increased levels of TLR4, DPPI, IL-6, TNF-α, MMP-2/9 in NEC ileum were suppressed by SHMOs in-vivo. SHMOs prevented Caco-2 cells from LPS, histamine, chymase induced damages by surviving cell viability, regulating G0/G1 and S phase in cell cycles, and increasing mitochondrial membrane potential. These findings provide a new insight into the pharmacological mechanism of SHMOs treatment for NEC and suggest that SHMOs needs well attention for therapeutic aims.


Assuntos
Catepsina C/metabolismo , Enterocolite Necrosante/prevenção & controle , Íleo/patologia , Mastócitos/metabolismo , Leite Humano/química , Oligossacarídeos/farmacologia , Receptor 4 Toll-Like/metabolismo , Animais , Animais Recém-Nascidos , Células CACO-2 , Ciclo Celular/efeitos dos fármacos , Modelos Animais de Doenças , Enterocolite Necrosante/imunologia , Enterocolite Necrosante/metabolismo , Glutationa/metabolismo , Humanos , Íleo/efeitos dos fármacos , Mastócitos/efeitos dos fármacos , Metaloproteinase 2 da Matriz/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Leite Humano/imunologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Ácido N-Acetilneuramínico/química , Ácido N-Acetilneuramínico/imunologia , Ácido N-Acetilneuramínico/farmacologia , Oligossacarídeos/química , Oligossacarídeos/imunologia , Ratos Sprague-Dawley
17.
Biochem Genet ; 58(6): 883-900, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32607676

RESUMO

Sialic acid (N-acetylneuraminic acid, NANA) is found at all cell surfaces of vertebrates. Although it is widely accepted that sialic acid is an essential substrate for brain development via a significant role in nerve transfers, structure of glycosides, and synaptogenesis phenomena, there are some reports on the elevated levels of sialic acid and prevalence of neurodegeneration. Matrix metalloproteases (MMPs) and tissue inhibitor of metalloproteinases (TIMPs) are involved in neuroinflammation disorders and produced by many cell types, including activated T cells, macrophages, neurons, astrocytes, and microglial cells. It can be hypothesized that sialic acid may have a potentially critical role in regulation of a wide range of uncovered neurodegeneration factors as its downstream targets. In this study, for the first time, we aimed to analyze the possible effect of the sialic acid solution exposure in the human C118 cell line, which was derived from a human brain astrocytoma (glial cells), on the expression patterns of miR-218, NF-kB, MMP-9, and TIMP-1. For MMP-9, protein levels were studied too. Half maximal inhibitory concentration (IC50) value of NANA was obtained by MTT assay. Glial cell line was treated with sialic acid (300, 500, and 1000 µg/ml) for 24 h to investigate the effects of this ligand on the expression of miR-218, NF-kB, MMP-9, and TIMP-1 genes. Protein levels were checked by Western blotting, and by using zymography, the gelatinolytic activity of MMP-9 secreted into conditioned media was assayed. At 300 µM, 500 µM, and 1000 µM sialic acid treatments, the expression of miR-218 was downregulated; subsequently, the NF-kB, MMP-9, and TIMP-1 genes as well as their protein expressions were upregulated. More interestingly, the enzyme activity of secreted MMP-9 was upregulated too (p-values ≤ 0.05). This study could demonstrate the significant effect of sialic acid on miR-218, NF-kB, MMP-9 , and TIMP-1 expressions in gene and protein levels and also the levels of enzyme activity of secreted MMP-9. Therefore, provided information indicates the novel idea of a possible linkage between sialic acid species and regulation of these neuroinflammation genes in Glial cell line.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Metaloproteinase 9 da Matriz/biossíntese , MicroRNAs/biossíntese , Ácido N-Acetilneuramínico/farmacologia , NF-kappa B/biossíntese , Neuroglia/metabolismo , Inibidor Tecidual de Metaloproteinase-1/biossíntese , Linhagem Celular Tumoral , Humanos
18.
Biomater Sci ; 8(2): 577-585, 2020 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-31872195

RESUMO

Hypersialylation is the aberrant expression of sialic acid in cell surface glycans and is pervasive in cancer cells. Recent studies have shown that hypersialylation provides a microenvironment conducive to cancer progression, mediated by the interaction between sialic acid and sialic acid-binding receptors. Therefore, a technique to block the interaction between the overexpressed sialic acid on cancer cell surfaces and its receptors is a promising approach to develop new cancer therapies. We focused on hydrogels as an artificial barrier to block this interaction and present here the development of a novel technique for selectively covalently binding a thin hydrogel barrier on sialic acid residues on cancer cell surfaces. This technique effectively inhibited cancer cell adhesion, motility and growth, caused cancer cell death in vitro, and completely suppressed tumor growth in vivo, thereby clearly demonstrating a potent antitumor effect.


Assuntos
Antineoplásicos/farmacologia , Membrana Celular/química , Hidrogéis/farmacologia , Ácido N-Acetilneuramínico/farmacologia , Antineoplásicos/química , Configuração de Carboidratos , Adesão Celular/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Hidrogéis/química , Células MCF-7 , Ácido N-Acetilneuramínico/química
19.
Nat Chem Biol ; 15(10): 949-958, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31451760

RESUMO

Antibody-drug conjugates (ADCs) selectively deliver chemotherapeutic agents to target cells and are important cancer therapeutics. However, the mechanisms by which ADCs are internalized and activated remain unclear. Using CRISPR-Cas9 screens, we uncover many known and novel endolysosomal regulators as modulators of ADC toxicity. We identify and characterize C18ORF8/RMC1 as a regulator of ADC toxicity through its role in endosomal maturation. Through comparative analysis of screens with ADCs bearing different linkers, we show that a subset of late endolysosomal regulators selectively influence toxicity of noncleavable linker ADCs. Surprisingly, we find cleavable valine-citrulline linkers can be processed rapidly after internalization without lysosomal delivery. Lastly, we show that sialic acid depletion enhances ADC lysosomal delivery and killing in diverse cancer cell types, including with FDA (US Food and Drug Administration)-approved trastuzumab emtansine (T-DM1) in Her2-positive breast cancer cells. Together, these results reveal new regulators of endolysosomal trafficking, provide important insights for ADC design and identify candidate combination therapy targets.


Assuntos
Sistemas CRISPR-Cas , Estudo de Associação Genômica Ampla , Imunoconjugados/toxicidade , Maitansina/análogos & derivados , Ácido N-Acetilneuramínico/farmacologia , Trastuzumab/farmacologia , Ado-Trastuzumab Emtansina , Antineoplásicos Imunológicos/farmacologia , Proteínas de Transporte , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Técnicas de Inativação de Genes , Humanos , Lisossomos , Maitansina/farmacologia
20.
J Biol Chem ; 294(12): 4437-4449, 2019 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-30670592

RESUMO

Neuroblastoma cells highly express the disialoganglioside GD2, a tumor-associated carbohydrate antigen, which is only sparsely expressed on healthy tissue. GD2 is a primary target for the development of immunotherapy for neuroblastoma. Immunotherapy with monoclonal anti-GD2 antibodies has proven safety and efficacy in clinical trials and is included in the standard treatment for children with high-risk neuroblastoma. Strategies to modulate GD2 expression in neuroblastoma could further improve anti-GD2-targeted immunotherapy. Here, we report that the cellular sialylation pathway, as well as epigenetic reprogramming, strongly modulates GD2 expression in human and mouse neuroblastoma cell lines. Recognition of GD2 by the 14G2a antibody is sialic acid-dependent and was blocked with the fluorinated sialic acid mimetic Ac53FaxNeu5Ac. Interestingly, sialic acid supplementation using a cell-permeable sialic acid analogue (Ac5Neu5Ac) boosted GD2 expression without or with minor alterations in overall cell surface sialylation. Furthermore, sialic acid supplementation with Ac5Neu5Ac combined with various histone deacetylase (HDAC) inhibitors, including vorinostat, enhanced GD2 expression in neuroblastoma cells beyond their individual effects. Mechanistic studies revealed that Ac5Neu5Ac supplementation increased intracellular CMP-Neu5Ac concentrations, thereby providing higher substrate levels for sialyltransferases. Furthermore, HDAC inhibitor treatment increased mRNA expression of the sialyltransferases GM3 synthase (ST3GAL5) and GD3 synthase (ST8SIA1), both of which are involved in GD2 biosynthesis. Our findings reveal that sialic acid analogues and HDAC inhibitors enhance GD2 expression and could potentially be employed to boost anti-GD2 targeted immunotherapy in neuroblastoma patients.


Assuntos
Antígenos de Neoplasias/metabolismo , Gangliosídeos/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Ácido N-Acetilneuramínico/farmacologia , Neuroblastoma/imunologia , Regulação para Cima/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Imunoterapia , Camundongos , Neuroblastoma/enzimologia , Neuroblastoma/patologia , Neuroblastoma/terapia , Sialiltransferases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA