Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 10.962
Filtrar
Más filtros











Intervalo de año de publicación
1.
Front Cell Infect Microbiol ; 14: 1394713, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38836054

RESUMEN

The rabies virus enters the nervous system by interacting with several molecular targets on host cells to modify behavior and trigger receptor-mediated endocytosis of the virion by poorly understood mechanisms. The rabies virus glycoprotein (RVG) interacts with the muscle acetylcholine receptor and the neuronal α4ß2 subtype of the nicotinic acetylcholine receptor (nAChR) family by the putative neurotoxin-like motif. Given that the neurotoxin-like motif is highly homologous to the α7 nAChR subtype selective snake toxin α-bungarotoxin (αBTX), other nAChR subtypes are likely involved. The purpose of this study is to determine the activity of the RVG neurotoxin-like motif on nAChR subtypes that are expressed in brain regions involved in rabid animal behavior. nAChRs were expressed in Xenopus laevis oocytes, and two-electrode voltage clamp electrophysiology was used to collect concentration-response data to measure the functional effects. The RVG peptide preferentially and completely inhibits α7 nAChR ACh-induced currents by a competitive antagonist mechanism. Tested heteromeric nAChRs are also inhibited, but to a lesser extent than the α7 subtype. Residues of the RVG peptide with high sequence homology to αBTX and other neurotoxins were substituted with alanine. Altered RVG neurotoxin-like peptides showed that residues phenylalanine 192, arginine 196, and arginine 199 are important determinants of RVG peptide apparent potency on α7 nAChRs, while serine 195 is not. The evaluation of the rabies ectodomain reaffirmed the observations made with the RVG peptide, illustrating a significant inhibitory impact on α7 nAChR with potency in the nanomolar range. In a mammalian cell culture model of neurons, we confirm that the RVG peptide binds preferentially to cells expressing the α7 nAChR. Defining the activity of the RVG peptide on nAChRs expands our understanding of basic mechanisms in host-pathogen interactions that result in neurological disorders.


Asunto(s)
Glicoproteínas , Virus de la Rabia , Xenopus laevis , Receptor Nicotínico de Acetilcolina alfa 7 , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Animales , Virus de la Rabia/fisiología , Virus de la Rabia/metabolismo , Humanos , Glicoproteínas/metabolismo , Glicoproteínas/genética , Oocitos/metabolismo , Proteínas Virales/metabolismo , Proteínas Virales/genética , Proteínas del Envoltorio Viral/metabolismo , Proteínas del Envoltorio Viral/genética , Interacciones Huésped-Patógeno , Unión Proteica , Rabia/metabolismo , Rabia/virología , Acetilcolina/metabolismo , Acetilcolina/farmacología , Neurotoxinas/metabolismo , Neurotoxinas/farmacología
2.
Methods Mol Biol ; 2804: 117-125, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38753144

RESUMEN

Several glycoproteins are validated biomarkers of various diseases such as cancer, cardiovascular diseases, chronic alcohol abuse, or congenital disorders of glycosylation (CDG). In particular, CDG represent a group of more than 150 inherited diseases with varied symptoms affecting multiple organs. The distribution of glycans from target glycoprotein(s) can be used to extract information to help the diagnosis and possibly differentiate subtypes of CDG. Indeed, depending on the glycans and the proteins to which they are attached, glycans can play a very broad range of roles in both physical and biological properties of glycoproteins. For glycans in general, capillary electrophoresis with laser-induced fluorescence detection (CE-LIF) has become a staple. Analysis of glycans with CE-LIF requires several sample preparation steps, including release of glycans from the target glycoprotein, fluorescent labeling of glycans, and purification of labeled glycans. Here, we describe the protocol for glycan sample treatment in a microfluidic droplet system prior to CE-LIF of labeled glycans. The microfluidic droplet approach offers full automation, sample, and reagent volume reduction and elimination of contamination from external environment.


Asunto(s)
Biomarcadores , Electroforesis Capilar , Polisacáridos , Electroforesis Capilar/métodos , Biomarcadores/análisis , Polisacáridos/análisis , Humanos , Glicoproteínas/análisis , Glicoproteínas/metabolismo , Microfluídica/métodos , Microfluídica/instrumentación , Glicosilación
3.
Pharmacol Res ; 204: 107218, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38768671

RESUMEN

This study investigates the role of Stanniocalcin-1 (STC1) in melanoma progression, with a focus on its impact on metastasis, angiogenesis, and immune evasion. Systematic bioinformatics analysis revealed the potential influence of STC1 dysregulation on prognosis, immune cell infiltration, response to immune therapy, and cellular functions. In vitro assays were conducted to assess the proliferation, invasion, migration, and angiogenesis capabilities of A375 cells. In vivo experiments utilizing C57BL/6 J mice established a lung metastasis model using B16-F10 cells to evaluate macrophage infiltration and M2 polarization. A Transwell co-culture system was employed to explore the crosstalk between melanoma and macrophages. Molecular interactions among STC1, YAP, ßPIX, and CCL2 are investigated using mass spectrometry, Co-Immunoprecipitation, Dual-Luciferase Reporter Assay, and Chromatin Immunoprecipitation experiments. STC1 was found to enhance lung metastasis by promoting the recruitment and polarization of M2 macrophages, thereby fostering an immunosuppressive microenvironment. Mechanistically, STC1 competes with YAP for binding to ßPIX within the KER domain in melanoma cells, leading to YAP activation and subsequent CCL2 upregulation. CCL2-induced M2 macrophages secrete VEGFA, which enhances tumor vascularization and increases STC1 expression via the AKT signaling pathway in melanoma cells, establishing a pro-metastatic feedback loop. Notably, STC1-induced YAP activation increases PD-L1 expression, promoting immune evasion. Silencing STC1 enhances the efficacy of PD-1 immune checkpoint therapy in mice. This research elucidates STC1's role in melanoma metastasis and its complex interactions with tumor-associated macrophages, proposing STC1 as a potential therapeutic target for countering melanoma metastasis and augmenting the efficacy of PD-1 immunotherapy.


Asunto(s)
Quimiocina CCL2 , Glicoproteínas , Macrófagos , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-akt , Factor A de Crecimiento Endotelial Vascular , Proteínas Señalizadoras YAP , Animales , Proteínas Señalizadoras YAP/metabolismo , Proteínas Señalizadoras YAP/genética , Humanos , Quimiocina CCL2/metabolismo , Quimiocina CCL2/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Línea Celular Tumoral , Macrófagos/metabolismo , Macrófagos/inmunología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Glicoproteínas/metabolismo , Glicoproteínas/genética , Ratones , Melanoma/patología , Melanoma/metabolismo , Melanoma/inmunología , Melanoma/genética , Retroalimentación Fisiológica , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Microambiente Tumoral , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Melanoma Experimental/metabolismo , Progresión de la Enfermedad , Transducción de Señal , Factores de Transcripción/metabolismo , Factores de Transcripción/genética
4.
Microbiol Spectr ; 12(6): e0034624, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38709084

RESUMEN

Across the Burkholderia genus O-linked protein glycosylation is highly conserved. While the inhibition of glycosylation has been shown to be detrimental for virulence in Burkholderia cepacia complex species, such as Burkholderia cenocepacia, little is known about how specific glycosylation sites impact protein functionality. Within this study, we sought to improve our understanding of the breadth, dynamics, and requirement for glycosylation across the B. cenocepacia O-glycoproteome. Assessing the B. cenocepacia glycoproteome across different culture media using complementary glycoproteomic approaches, we increase the known glycoproteome to 141 glycoproteins. Leveraging this repertoire of glycoproteins, we quantitively assessed the glycoproteome of B. cenocepacia using Data-Independent Acquisition (DIA) revealing the B. cenocepacia glycoproteome is largely stable across conditions with most glycoproteins constitutively expressed. Examination of how the absence of glycosylation impacts the glycoproteome reveals that the protein abundance of only five glycoproteins (BCAL1086, BCAL2974, BCAL0525, BCAM0505, and BCAL0127) are altered by the loss of glycosylation. Assessing ΔfliF (ΔBCAL0525), ΔmotB (ΔBCAL0127), and ΔBCAM0505 strains, we demonstrate the loss of FliF, and to a lesser extent MotB, mirror the proteomic effects observed in the absence of glycosylation in ΔpglL. While both MotB and FliF are essential for motility, we find loss of glycosylation sites in MotB or FliF does not impact motility supporting these sites are dispensable for function. Combined this work broadens our understanding of the B. cenocepacia glycoproteome supporting that the loss of glycoproteins in the absence of glycosylation is not an indicator of the requirement for glycosylation for protein function. IMPORTANCE: Burkholderia cenocepacia is an opportunistic pathogen of concern within the Cystic Fibrosis community. Despite a greater appreciation of the unique physiology of B. cenocepacia gained over the last 20 years a complete understanding of the proteome and especially the O-glycoproteome, is lacking. In this study, we utilize systems biology approaches to expand the known B. cenocepacia glycoproteome as well as track the dynamics of glycoproteins across growth phases, culturing media and in response to the loss of glycosylation. We show that the glycoproteome of B. cenocepacia is largely stable across conditions and that the loss of glycosylation only impacts five glycoproteins including the motility associated proteins FliF and MotB. Examination of MotB and FliF shows, while these proteins are essential for motility, glycosylation is dispensable. Combined this work supports that B. cenocepacia glycosylation can be dispensable for protein function and may influence protein properties beyond stability.


Asunto(s)
Proteínas Bacterianas , Burkholderia cenocepacia , Glicoproteínas , Proteómica , Glicosilación , Burkholderia cenocepacia/metabolismo , Burkholderia cenocepacia/genética , Burkholderia cenocepacia/crecimiento & desarrollo , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Glicoproteínas/metabolismo , Glicoproteínas/genética , Proteoma/metabolismo
5.
Appl Microbiol Biotechnol ; 108(1): 303, 2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38639795

RESUMEN

Severe fever with thrombocytopenia syndrome virus (SFTSV) causes the highly fatal disease in humans. To facilitate diagnosis, the native form of subunit glycoprotein (Gn), a prime target for potential vaccines and therapies, was produced in Nicotiana benthamiana using a Bamboo mosaic virus-based vector system. By fusion with secretory signal tags, SSExt, derived from the extension protein, and the (SP)10 motif, the yield of the recombinant Gn (rGn) was remarkably increased to approximately 7 mg/kg infiltrated leaves. Ultimately, an rGn-based ELISA was successfully established for the detection of SFTSV-specific antibodies in serum samples from naturally infected monkeys. As validated with the reference method, the specificity and sensitivity of rGn-ELISA were 94% and 96%, respectively. In conclusion, utilizing well-suited fusion tags facilitates rGn production and purification in substantial quantities while preserving its antigenic properties. The rGn-ELISA, characterized by its commendable sensitivity and specificity could serve as a viable alternative diagnostic method for assessing SFTSV seroprevalence. KEY POINTS: • SFTSV Gn, fused with secretory signal tags, was expressed by the BaMV-based vector. • The plant fusion tags increased expression levels and eased the purification of rGn. • The rGn-ELISA was established and validated; its specificity and sensitivity > 94%.


Asunto(s)
Phlebovirus , Síndrome de Trombocitopenia Febril Grave , Humanos , Síndrome de Trombocitopenia Febril Grave/diagnóstico , Phlebovirus/genética , Phlebovirus/metabolismo , Estudios Seroepidemiológicos , Glicoproteínas/metabolismo , Anticuerpos
6.
Glycobiology ; 34(6)2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38590172

RESUMEN

Human noroviruses, globally the main cause of viral gastroenteritis, show strain specific affinity for histo-blood group antigens (HBGA) and can successfully be propagated ex vivo in human intestinal enteroids (HIEs). HIEs established from jejunal stem cells of individuals with different ABO, Lewis and secretor geno- and phenotypes, show varying susceptibility to such infections. Using bottom-up glycoproteomic approaches we have defined and compared the N-linked glycans of glycoproteins of seven jejunal HIEs. Membrane proteins were extracted, trypsin digested, and glycopeptides enriched by hydrophilic interaction liquid chromatography and analyzed by nanoLC-MS/MS. The Byonic software was used for glycopeptide identification followed by hands-on verifications and interpretations. Glycan structures and attachment sites were identified from MS2 spectra obtained by higher-energy collision dissociation through analysis of diagnostic saccharide oxonium ions (B-ions), stepwise glycosidic fragmentation of the glycans (Y-ions), and peptide sequence ions (b- and y-ions). Altogether 694 unique glycopeptides from 93 glycoproteins were identified. The N-glycans encompassed pauci- and oligomannose, hybrid- and complex-type structures. Notably, polyfucosylated HBGA-containing glycopeptides of the four glycoproteins tetraspanin-8, carcinoembryonic antigen-related cell adhesion molecule 5, sucrose-isomaltase and aminopeptidase N were especially prominent and were characterized in detail and related to donor ABO, Lewis and secretor types of each HIE. Virtually no sialylated N-glycans were identified for these glycoproteins suggesting that terminal sialylation was infrequent compared to fucosylation and HBGA biosynthesis. This approach gives unique site-specific information on the structural complexity of N-linked glycans of glycoproteins of human HIEs and provides a platform for future studies on the role of host glycoproteins in gastrointestinal infectious diseases.


Asunto(s)
Glicoproteínas , Humanos , Glicoproteínas/metabolismo , Glicoproteínas/química , Proteómica/métodos , Antígenos de Grupos Sanguíneos/metabolismo , Antígenos de Grupos Sanguíneos/química , Polisacáridos/química , Polisacáridos/metabolismo , Fucosa/metabolismo , Fucosa/química , Fenotipo , Glicosilación , Sistema del Grupo Sanguíneo ABO/metabolismo , Sistema del Grupo Sanguíneo ABO/química
7.
Cell Rep Methods ; 4(4): 100744, 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38582075

RESUMEN

A comprehensive analysis of site-specific protein O-glycosylation is hindered by the absence of a consensus O-glycosylation motif, the diversity of O-glycan structures, and the lack of a universal enzyme that cleaves attached O-glycans. Here, we report the development of a robust O-glycoproteomic workflow for analyzing complex biological samples by combining four different strategies: removal of N-glycans, complementary digestion using O-glycoprotease (IMPa) with/without another protease, glycopeptide enrichment, and mass spectrometry with fragmentation of glycopeptides using stepped collision energy. Using this workflow, we cataloged 474 O-glycopeptides on 189 O-glycosites derived from 79 O-glycoproteins from human plasma. These data revealed O-glycosylation of several abundant proteins that have not been previously reported. Because many of the proteins that contained unannotated O-glycosylation sites have been extensively studied, we wished to confirm glycosylation at these sites in a targeted fashion. Thus, we analyzed selected purified proteins (kininogen-1, fetuin-A, fibrinogen, apolipoprotein E, and plasminogen) in independent experiments and validated the previously unknown O-glycosites.


Asunto(s)
Glicoproteínas , Proteoma , Proteómica , Flujo de Trabajo , Humanos , Glicosilación , Glicoproteínas/metabolismo , Glicoproteínas/química , Proteómica/métodos , Proteoma/metabolismo , Proteoma/análisis , Glicopéptidos/análisis , Glicopéptidos/química , Glicopéptidos/metabolismo , Quininógenos/metabolismo , Quininógenos/química , Polisacáridos/metabolismo , Apolipoproteínas E/metabolismo , Apolipoproteínas E/química , Fibrinógeno/metabolismo , Fibrinógeno/química , alfa-2-Glicoproteína-HS/metabolismo , alfa-2-Glicoproteína-HS/análisis
8.
Int J Biol Macromol ; 266(Pt 2): 131341, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38574922

RESUMEN

Sialic acids are negatively charged carbohydrates that are components of saccharide chains covalently linked to macromolecules. Sialylated glycoproteins are important for most biological processes, including reproduction, where they are associated with spermatogenesis, sperm motility, immune responses, and fertilization. Changes in the glycoprotein profile or sialylation in glycoproteins are likely to affect the quality of ejaculate. The aim of this study was to determine differences in the degree of sialylation between normozoospermic ejaculates and ejaculates with a pathological spermiogram using two lectins, Sambucus nigra (SNA) and Maackia amurensis (MAL II/MAA) recognizing α-2,6 or α-2,3 linkage of Sia to galactosyl residues. Our results show a close relationship between seminal plasma (SP) sialoproteins and the presence of anti-sperm antibodies in the ejaculate, apoptotic spermatozoa, and ejaculate quality. Using mass spectrometry, we identified SP sialoproteins such as, semenogelins, glycodelin, prolactin-inducible protein, lactotransferrin, and clusterin that are associated with spermatozoa and contribute to the modulation of the immune response and sperm apoptosis. Our findings suggest a correlation between the degree of SP glycoprotein sialylation and the existence of possible pathological states of spermatozoa and reproductive organs. Glycoproteins sialylation represents a potential parameter reflecting the overall quality of ejaculate and could potentially be utilised in diagnostics.


Asunto(s)
Semen , Espermatozoides , Masculino , Humanos , Semen/metabolismo , Semen/química , Espermatozoides/metabolismo , Motilidad Espermática , Glicoproteínas/metabolismo , Glicodelina/metabolismo , Proteínas de Secreción de la Vesícula Seminal/metabolismo , Análisis de Semen/métodos , Clusterina/metabolismo , Lectinas/metabolismo , Lectinas/química , Eyaculación , Ácidos Siálicos/metabolismo , Proteínas de Plasma Seminal/metabolismo , Lactoferrina/metabolismo , Apoptosis
9.
J Transl Med ; 22(1): 383, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38659028

RESUMEN

BACKGROUND: Loss of AZGP1 expression is a biomarker associated with progression to castration resistance, development of metastasis, and poor disease-specific survival in prostate cancer. However, high expression of AZGP1 cells in prostate cancer has been reported to increase proliferation and invasion. The exact role of AZGP1 in prostate cancer progression remains elusive. METHOD: AZGP1 knockout and overexpressing prostate cancer cells were generated using a lentiviral system. The effects of AZGP1 under- or over-expression in prostate cancer cells were evaluated by in vitro cell proliferation, migration, and invasion assays. Heterozygous AZGP1± mice were obtained from European Mouse Mutant Archive (EMMA), and prostate tissues from homozygous knockout male mice were collected at 2, 6 and 10 months for histological analysis. In vivo xenografts generated from AZGP1 under- or over-expressing prostate cancer cells were used to determine the role of AZGP1 in prostate cancer tumor growth, and subsequent proteomics analysis was conducted to elucidate the mechanisms of AZGP1 action in prostate cancer progression. AZGP1 expression and microvessel density were measured in human prostate cancer samples on a tissue microarray of 215 independent patient samples. RESULT: Neither the knockout nor overexpression of AZGP1 exhibited significant effects on prostate cancer cell proliferation, clonal growth, migration, or invasion in vitro. The prostates of AZGP1-/- mice initially appeared to have grossly normal morphology; however, we observed fibrosis in the periglandular stroma and higher blood vessel density in the mouse prostate by 6 months. In PC3 and DU145 mouse xenografts, over-expression of AZGP1 did not affect tumor growth. Instead, these tumors displayed decreased microvessel density compared to xenografts derived from PC3 and DU145 control cells, suggesting that AZGP1 functions to inhibit angiogenesis in prostate cancer. Proteomics profiling further indicated that, compared to control xenografts, AZGP1 overexpressing PC3 xenografts are enriched with angiogenesis pathway proteins, including YWHAZ, EPHA2, SERPINE1, and PDCD6, MMP9, GPX1, HSPB1, COL18A1, RNH1, and ANXA1. In vitro functional studies show that AZGP1 inhibits human umbilical vein endothelial cell proliferation, migration, tubular formation and branching. Additionally, tumor microarray analysis shows that AZGP1 expression is negatively correlated with blood vessel density in human prostate cancer tissues. CONCLUSION: AZGP1 is a negative regulator of angiogenesis, such that loss of AZGP1 promotes angiogenesis in prostate cancer. AZGP1 likely exerts heterotypical effects on cells in the tumor microenvironment, such as stromal and endothelial cells. This study sheds light on the anti-angiogenic characteristics of AZGP1 in the prostate and provides a rationale to target AZGP1 to inhibit prostate cancer progression.


Asunto(s)
Movimiento Celular , Proliferación Celular , Neovascularización Patológica , Neoplasias de la Próstata , Masculino , Animales , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Humanos , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Línea Celular Tumoral , Ratones Noqueados , Glicoproteínas/metabolismo , Invasividad Neoplásica , Ratones , Regulación Neoplásica de la Expresión Génica , Angiogénesis , Zn-alfa-2-Glicoproteína
10.
Int J Biol Macromol ; 268(Pt 1): 131511, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38615867

RESUMEN

This comprehensive review delves into the intricate landscape of glycans and glycoconjugates, unraveling their multifaceted roles across diverse biological dimensions. From influencing fundamental cellular processes such as signaling, recognition, and adhesion to exerting profound effects at the molecular and genetic levels, these complex carbohydrate structures emerge as linchpins in cellular functions and interactions. The structural diversity of glycoconjugates, which can be specifically classified into glycoproteins, glycolipids, and proteoglycans, underscores their importance in shaping the architecture of cells. Beyond their structural roles, these molecules also play key functions in facilitating cellular communication and modulating recognition mechanisms. Further, glycans and glycoconjugates prove invaluable as biomarkers in disease diagnostics, particularly in cancer, where aberrant glycosylation patterns offer critical diagnostic cues. Furthermore, the review explores their promising therapeutic applications, ranging from the development of glycan-based nanomaterials for precise drug delivery to innovative interventions in cancer treatment. This review endeavors to comprehensively explore the intricate functions of glycans and glycoconjugates, with the primary goal of offering valuable insights into their extensive implications in both health and disease. Encompassing a broad spectrum of biological processes, the focus of the review aims to provide a comprehensive understanding of the significant roles played by glycans and glycoconjugates.


Asunto(s)
Glicoconjugados , Polisacáridos , Humanos , Polisacáridos/química , Polisacáridos/metabolismo , Glicoconjugados/química , Glicoconjugados/metabolismo , Animales , Neoplasias/metabolismo , Glicosilación , Glicoproteínas/química , Glicoproteínas/metabolismo
11.
J Nutr ; 154(6): 1790-1802, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38636707

RESUMEN

BACKGROUND: Stanniocalcin 2 (STC2), a glycoprotein hormone, is extensively expressed in various organs and tissues, particularly in the mammary gland. STC2 plays a crucial role in enabling cells to adapt to stress conditions and avert apoptosis. The efficiency of milk production is closely linked to both the quantity and quality of mammary cells. Yet, there remains a dearth of research on the impact of STC2 on mammary cells' activity in dairy cows. OBJECTIVES: The objective of this study was to investigate the effects of STC2 on the viability of mammary epithelial cells in dairy cows and to elucidate the underlying mechanisms. METHODS: First, the Gene Expression Profiling and Interactive Analysis database was employed to perform survival analysis on STC2 expression in relation to prognosis using The Cancer Genome Atlas and GETx data. Subsequently, the basic physical and chemical properties, gene expression, and potential signaling pathways involved in the growth of dairy cow mammary epithelial cells were determined using STC2 knockdown. RESULTS: STC2 knockdown significantly suppressed autophagy in mammary epithelial cells of dairy cows. Moreover, STC2 knockdown upregulated glutathione peroxidase 4 protein expression, elicited an elevation in lipid ROS concentrations, and inhibited the mechanistic target of rapamycin complex 1 (mTORC1) signaling pathway, consequently repressing downstream genes involved in lipid synthesis regulated by mTORC1 and ultimately inducing ferroptosis. CONCLUSIONS: The findings of our study suggest that STC2 suppresses autophagy and ferroptosis through the activation of mTORC1. Mechanically, STC2 exerts an inhibitory effect on ferroptosis by activating antioxidative stress-related proteins, such as glutathione peroxidase 4, to suppress lipid ROS production and stimulating the mTORC1 signaling pathway to enhance the expression of genes associated with lipid synthesis.


Asunto(s)
Autofagia , Células Epiteliales , Ferroptosis , Glicoproteínas , Glándulas Mamarias Animales , Diana Mecanicista del Complejo 1 de la Rapamicina , Animales , Bovinos , Femenino , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/citología , Células Epiteliales/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ferroptosis/efectos de los fármacos , Ferroptosis/fisiología , Glicoproteínas/metabolismo , Glicoproteínas/genética , Transducción de Señal
12.
FASEB J ; 38(6): e23551, 2024 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-38489235

RESUMEN

Inflammation is a significant pathological manifestation of inflammatory bowel disease (IBD), yet its mechanism has remained unclear. Although WNT2B is enriched in the intestinal inflammatory tissue of IBD patients, the specific mechanism of WNT2B in the formation of intestinal inflammation remains unclear. This study was aimed to investigate whether macrophages expressing WNT2B can aggravate intestinal tissue inflammation. Samples were collected from both normal individuals and patients with IBD at multiple colon sites. Macrophages were identified using tissue immunofluorescence. IκB kinase (IKK)-interacting protein (IKIP), which interacts with WNT2B, was found by protein cross-linking and protein mass spectrometry. The expression of WNT2B, IKIP, the NF-κB pathway, and downstream molecules were analyzed. An acute colitis model of C57BL/6J mice was established using an adeno-associated virus (AAV)-mediated WNT2B knockdown system and 3% dextran sulfate sodium (DSS). The degree of intestinal inflammation in mice was assessed upon WNT2B knockdown in macrophages. Macrophages expressing WNT2B were found to be enriched in the colitis tissues of IBD patients. WNT2B in macrophages activated the NF-κB pathway and enhanced the expression of downstream inflammatory cytokines. By competitively binding IKIP, WNT2B reduced the binding of IKIP to IKKß and promoted the activation of the NF-κB pathway. Using an AAV-mediated WNT2B knockdown system, WNT2B expression in intestinal macrophages was suppressed, leading to a reduction in intestinal inflammation. WNT2B activated the NF-κB pathway and enhanced the expression of downstream inflammatory cytokines by competitively binding to IKIP, potentially contributing to colon inflammatory injury in IBD.


Asunto(s)
Colitis , Enfermedades Inflamatorias del Intestino , Humanos , Ratones , Animales , FN-kappa B/metabolismo , Ratones Endogámicos C57BL , Transducción de Señal , Enfermedades Inflamatorias del Intestino/metabolismo , Colitis/metabolismo , Inflamación/metabolismo , Citocinas/metabolismo , Macrófagos/metabolismo , Sulfato de Dextran , Glicoproteínas/metabolismo , Proteínas Wnt/metabolismo
13.
Neuroreport ; 35(6): 374-379, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38526932

RESUMEN

Lethal giant larvae 1 (LGL1) is originally recognized as a tumor suppressor, implicated in maintaining cell polarity in Drosophila and mammalian cells. Cell polarity plays a crucial role in tumorigenesis. We previously established Pax2-LGL1 -/- conditional knockout mice but did not focus on the tumorigenesis in cerebellar primordium. HE staining was used to detect the morphological structure of the cerebellar primordium during early embryonic development in Pax2-LGL1 -/- mice. Immunofluorescence assays were used to detect the expression of polar molecules. TUNEL staining assessed tissue apoptosis. Our findings reveal that deletion of LGL1 leads to the emergence of neuroblastoma-like tissues within the cerebellum primordium during early embryogenesis. This outcome can be attributed to alterations in expression patterns of polar molecules Cdc42 and ß-catenin following early deletion of LGL1, resulting in loss of cell polarity among neuroepithelial cells and subsequent formation of tumor-like tissues. However, further histological examination demonstrated that these tumor-like tissues disappear from embryonic day 15.5 onwards within the cerebellar primordium of Pax2-LGL1 -/- mice due to apoptosis-mediated cellular compensation. Our data emphasize the importance of LGL1 in maintaining neuroepithelial cell polarity and reveal a novel role for LGL1 in regulating tumorigenesis and ablation in the cerebellar primordium.


Asunto(s)
Apoptosis , Cerebelo , Glicoproteínas , Animales , Ratones , Carcinogénesis , Cerebelo/metabolismo , Glicoproteínas/metabolismo , Mamíferos/metabolismo , Ratones Noqueados
14.
Virology ; 594: 110059, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38518442

RESUMEN

Ovine pulmonary adenocarcinoma (OPA), caused by the jaagsiekte sheep retrovirus (JSRV), is a chronic, progressive, and contagious lung tumor that seriously affects sheep production. It also represents a valuable animal model for several human lung adenocarcinomas. However, little is known about the role of autophagy in OPA tumorigenesis. Here, Western blotting combined with transmission electron microscopy examination and Cyto-ID dye staining was employed for evaluation of changes of autophagic levels. The results of the present study showed that expression of the autophagy marker proteins Beclin-1 and LC3 was decreased in OPA lung tissues, as well as in cells overexpressing the envelope glycoprotein of JSRV (JSRV Env). Reduced numbers of autophagosomes were also observed in cells overexpressing JSRV Env, although assessment of autophagic flux showed that JSRV Env overexpression did not block the formation of autophagosomes, suggesting increased degradation of autolysosomes. Last, mouse xenograft experiments indicated that inhibition of autophagy by 3-methyladenine suppressed both tumor growth and the epithelial-to-mesenchymal transition. In conclusion, JSRV, through JSRV Env, takes advantage of the autophagy process, leading to the development of OPA.


Asunto(s)
Retrovirus Ovino Jaagsiekte , Ovinos , Animales , Humanos , Ratones , Retrovirus Ovino Jaagsiekte/genética , Retrovirus Ovino Jaagsiekte/metabolismo , Productos del Gen env , Transformación Celular Neoplásica , Autofagia , Glicoproteínas/metabolismo
15.
J Dent Res ; 103(6): 631-641, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38491721

RESUMEN

Periodontal tissue destruction in periodontitis is a consequence of the host inflammatory response to periodontal pathogens, which could be aggravated in the presence of type 2 diabetes mellitus (T2DM). Accumulating evidence highlights the intricate involvement of macrophage-mediated inflammation in the pathogenesis of periodontitis under both normal and T2DM conditions. However, the underlying mechanism remains elusive. Alpha-2-glycoprotein 1 (AZGP1), a glycoprotein featuring an MHC-I domain, has been implicated in both inflammation and metabolic disorders. In this study, we found that AZGP1 was primarily colocalized with macrophages in periodontitis tissues. AZGP1 was increased in periodontitis compared with controls, which was further elevated when accompanied by T2DM. Adeno-associated virus-mediated overexpression of Azgp1 in the periodontium significantly enhanced periodontal inflammation and alveolar bone loss, accompanied by elevated M1 macrophages and pyroptosis in murine models of periodontitis and T2DM-associated periodontitis, while Azgp1-/- mice exhibited opposite effects. In primary bone marrow-derived macrophages stimulated by lipopolysaccharide (LPS) or LPS and palmitic acid (PA), overexpression or knockout of Azgp1 markedly upregulated or suppressed, respectively, the expression of macrophage M1 markers and key components of the NLR Family Pyrin Domain Containing 3 (NLRP3)/caspase-1 signaling. Moreover, conditioned medium from Azgp1-overexpressed macrophages under LPS or LPS+PA stimulation induced higher inflammatory activation and lower osteogenic differentiation in human periodontal ligament stem cells (hPDLSCs). Furthermore, elevated M1 polarization and pyroptosis in macrophages and associated detrimental effects on hPDLSCs induced by Azgp1 overexpression could be rescued by NLRP3 or caspase-1 inhibition. Collectively, our study elucidated that AZGP1 could aggravate periodontitis by promoting macrophage M1 polarization and pyroptosis through the NLRP3/casapse-1 pathway, which was accentuated in T2DM-associated periodontitis. This finding deepens the understanding of AZGP1 in the pathogenesis of periodontitis and suggests AZGP1 as a crucial link mediating the adverse effects of diabetes on periodontal inflammation.


Asunto(s)
Diabetes Mellitus Tipo 2 , Macrófagos , Periodontitis , Piroptosis , Animales , Macrófagos/metabolismo , Periodontitis/metabolismo , Periodontitis/inmunología , Ratones , Humanos , Diabetes Mellitus Tipo 2/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Caspasa 1/metabolismo , Masculino , Ratones Noqueados , Transducción de Señal , Pérdida de Hueso Alveolar/metabolismo , Glicoproteínas/metabolismo
16.
J Hematol Oncol ; 17(1): 12, 2024 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-38515194

RESUMEN

Cancer early detection and treatment response prediction continue to pose significant challenges. Cancer liquid biopsies focusing on detecting circulating tumor cells (CTCs) and DNA (ctDNA) have shown enormous potential due to their non-invasive nature and the implications in precision cancer management. Recently, liquid biopsy has been further expanded to profile glycoproteins, which are the products of post-translational modifications of proteins and play key roles in both normal and pathological processes, including cancers. The advancements in chemical and mass spectrometry-based technologies and artificial intelligence-based platforms have enabled extensive studies of cancer and organ-specific changes in glycans and glycoproteins through glycomics and glycoproteomics. Glycoproteomic analysis has emerged as a promising tool for biomarker discovery and development in early detection of cancers and prediction of treatment efficacy including response to immunotherapies. These biomarkers could play a crucial role in aiding in early intervention and personalized therapy decisions. In this review, we summarize the significant advance in cancer glycoproteomic biomarker studies and the promise and challenges in integration into clinical practice to improve cancer patient care.


Asunto(s)
Inteligencia Artificial , Neoplasias , Humanos , Neoplasias/diagnóstico , Biomarcadores de Tumor/análisis , Glicoproteínas/análisis , Glicoproteínas/metabolismo , Biopsia Líquida , Proteoma
17.
Immunity ; 57(3): 559-573.e6, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38479361

RESUMEN

Epstein-Barr virus (EBV) causes infectious mononucleosis and is associated with B cell lymphomas. EBV glycoprotein 42 (gp42) binds HLA class II and activates membrane fusion with B cells. We isolated gp42-specific monoclonal antibodies (mAbs), A10 and 4C12, which use distinct mechanisms to neutralize virus infection. mAb A10 was more potent than the only known neutralizing gp42 mAb, F-2-1, in neutralizing EBV infection and blocking binding to HLA class II. mAb 4C12 was similar to mAb A10 in inhibiting glycoprotein-mediated B cell fusion but did not block receptor binding, and it was less effective in neutralizing infection. Crystallographic structures of gH/gL/gp42/A10 and gp42/4C12 complexes revealed two distinct sites of vulnerability on gp42 for receptor binding and B cell fusion. Passive transfer of mAb A10 into humanized mice conferred nearly 100% protection from viremia and EBV lymphomas after EBV challenge. These findings identify vulnerable sites on EBV that may facilitate therapeutics and vaccines.


Asunto(s)
Bencenoacetamidas , Infecciones por Virus de Epstein-Barr , Herpesvirus Humano 4 , Piperidonas , Animales , Ratones , Proteínas Virales/metabolismo , Glicoproteínas/metabolismo , Anticuerpos Antivirales
18.
Crit Rev Oncol Hematol ; 197: 104330, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38556071

RESUMEN

Sialic acid (SA), as the ultimate epitope of polysaccharides, can act as a cap at the end of polysaccharide chains to prevent their overextension. Sialylation is the enzymatic process of transferring SA residues onto polysaccharides and is catalyzed by a group of enzymes known as sialyltransferases (SiaTs). It is noteworthy that the sialylation level of glycoproteins is significantly altered when digestive cancer occurs. And this alteration exhibits a close correlation with the progression of these cancers. In this review, from the perspective of altered SiaTs expression levels and changed glycoprotein sialylation patterns, we summarize the pathogenesis of gastric cancer (GC), colorectal cancer (CRC), pancreatic ductal adenocarcinoma (PDAC), and hepatocellular carcinoma (HCC). Furthermore, we propose potential early diagnostic biomarkers and prognostic indicators for different digestive cancers. Finally, we summarize the therapeutic value of sialylation in digestive system cancers.


Asunto(s)
Biomarcadores de Tumor , Glicoproteínas , Sialiltransferasas , Humanos , Sialiltransferasas/metabolismo , Biomarcadores de Tumor/metabolismo , Glicoproteínas/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Neoplasias del Sistema Digestivo/metabolismo , Neoplasias del Sistema Digestivo/diagnóstico , Terapia Molecular Dirigida/métodos , Animales
19.
Adv Sci (Weinh) ; 11(21): e2309305, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38509833

RESUMEN

Spinal cord injury (SCI) has no effective treatment modalities. It faces a significant global therapeutical challenge, given its features of poor axon regeneration, progressive local inflammation, and inefficient systemic drug delivery due to the blood-spinal cord barrier (BSCB). To address these challenges, a new nano complex that achieves targeted drug delivery to the damaged spinal cord is proposed, which contains a mesoporous silica nanoparticle core loaded with microRNA and a cloaking layer of human umbilical cord mesenchymal stem cell membrane modified with rabies virus glycoprotein (RVG). The nano complex more readily crosses the damaged BSCB with its exosome-resembling properties, including appropriate size and a low-immunogenic cell membrane disguise and accumulates in the injury center because of RVG, where it releases abundant microRNAs to elicit axon sprouting and rehabilitate the inflammatory microenvironment. Culturing with nano complexes promotes axonal growth in neurons and M2 polarization in microglia. Furthermore, it showed that SCI mice treated with this nano complex by tail vein injection display significant improvement in axon regrowth, microenvironment regulation, and functional restoration. The efficacy and biocompatibility of the targeted delivery of microRNA by nano complexes demonstrate their immense potential as a noninvasive treatment for SCI.


Asunto(s)
Modelos Animales de Enfermedad , MicroARNs , Virus de la Rabia , Dióxido de Silicio , Traumatismos de la Médula Espinal , Animales , MicroARNs/genética , MicroARNs/administración & dosificación , Traumatismos de la Médula Espinal/terapia , Ratones , Dióxido de Silicio/química , Virus de la Rabia/genética , Glicoproteínas/metabolismo , Humanos , Células Madre Mesenquimatosas/metabolismo , Membrana Celular/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Nanopartículas/química
20.
Mol Carcinog ; 63(5): 938-950, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38353288

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is a highly invasive cancer with a poor prognosis and a 5-year survival rate of less than 11%. As a member of the CAP superfamily of proteins, the role of peptidase inhibitor 16 (Pi16) in tumor progression is still unclear. Immunohistochemistry and quantitative RT-PCR methods were used to detect the expression levels of Pi16 protein and mRNA in PDAC patients. CRISPR/Cas9 technology was used to knock out the expression of Pi16 in PDAC cell lines. In vivo and in vitro experiments were used to verify the effect of Pi16 on PDAC proliferation ability. By RNA sequencing, we found that oligoadenylate synthetase L (OASL) can serve as a potential downstream target of Pi16. The expression of Pi16 was higher in PDAC tissues than in matched adjacent tissues. High expression of Pi16 was associated with PDAC progression and poor prognosis. Overexpression of Pi16 could promote the proliferation of PDAC cells in vitro and in vivo. Bioinformatics analysis and coimmunoprecipitation assays showed that Pi16 could bind to OASL. Moreover, the functional recovery test confirmed that Pi16 could promote the proliferation of PDAC via OASL. Our present study demonstrates that Pi16 might participate in the occurrence and development of PDAC by regulating cell proliferation by binding to OASL, indicating that Pi16 might be a promising novel therapeutic target for PDAC.


Asunto(s)
2',5'-Oligoadenilato Sintetasa , Nucleótidos de Adenina , Carcinoma Ductal Pancreático , Glicoproteínas , Neoplasias Pancreáticas , Humanos , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Glicoproteínas/metabolismo , Proteínas Portadoras/metabolismo , 2',5'-Oligoadenilato Sintetasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA