Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 566
Filtrar
1.
Cell Mol Life Sci ; 81(1): 350, 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-39141086

RESUMEN

Heparan sulfate (HS) proteoglycans are important regulators of cellular responses to soluble mediators such as chemokines, cytokines and growth factors. We profiled changes in expression of genes encoding HS core proteins, biosynthesis enzymes and modifiers during macrophage polarisation, and found that the most highly regulated gene was Sulf2, an extracellular HS 6-O-sulfatase that was markedly downregulated in response to pro-inflammatory stimuli. We then generated Sulf2+/- bone marrow chimeric mice and examined inflammatory responses in antigen-induced arthritis, as a model of rheumatoid arthritis. Resolution of inflammation was impaired in myeloid Sulf2+/- chimeras, with elevated joint swelling and increased abundance of pro-arthritic Th17 cells in synovial tissue. Transcriptomic and in vitro analyses indicated that Sulf2 deficiency increased type I interferon signaling in bone marrow-derived macrophages, leading to elevated expression of the Th17-inducing cytokine IL6. This establishes that dynamic remodeling of HS by Sulf2 limits type I interferon signaling in macrophages, and so protects against Th17-driven pathology.


Asunto(s)
Macrófagos , Ratones Endogámicos C57BL , Transducción de Señal , Células Th17 , Animales , Células Th17/inmunología , Células Th17/metabolismo , Ratones , Macrófagos/metabolismo , Macrófagos/inmunología , Sulfatasas/metabolismo , Sulfatasas/genética , Sulfotransferasas/metabolismo , Sulfotransferasas/genética , Células Mieloides/metabolismo , Células Mieloides/inmunología , Artritis Experimental/inmunología , Artritis Experimental/patología , Artritis Experimental/metabolismo , Artritis Reumatoide/inmunología , Artritis Reumatoide/metabolismo , Artritis Reumatoide/patología , Inflamación/metabolismo , Inflamación/patología , Ratones Noqueados , Interleucina-6/metabolismo , Interleucina-6/genética , Heparitina Sulfato/metabolismo
2.
Mol Cell Proteomics ; 23(7): 100793, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38825040

RESUMEN

Human extracellular 6-O-endosulfatases Sulf-1 and Sulf-2 are the only enzymes that post-synthetically alter the 6-O sulfation of heparan sulfate proteoglycans (HSPG), which regulates interactions of HSPG with many proteins. Oncogenicity of Sulf-2 in different cancers has been documented, and we have shown that Sulf-2 is associated with poor survival outcomes in head and neck squamous cell carcinoma (HNSCC). Despite its importance, limited information is available on direct protein-protein interactions of the Sulf-2 protein in the tumor microenvironment. In this study, we used monoclonal antibody (mAb) affinity purification and mass spectrometry to identify galectin-3-binding protein (LG3BP) as a highly specific binding partner of Sulf-2 in the conditioned media of HNSCC cell lines. We validated their direct interaction in vitro using recombinant proteins and have shown that the chondroitin sulfate (CS) covalently bound to the Sulf-2 influences the binding to LG3BP. We confirmed the importance of the CS chain for the interaction by generating a mutant Sulf-2 protein that lacks the CS. Importantly, we have shown that the LG3BP inhibits Sulf-2 activity in vitro in a concentration-dependent manner. As a consequence, the addition of LG3BP to a spheroid cell culture inhibited the invasion of the HNSCC cells into Matrigel. Thus, Sulf-2 interaction with LG3BP may regulate the physiological activity of the Sulf-2 enzyme as well as its activity in the tumor microenvironment.


Asunto(s)
Unión Proteica , Sulfotransferasas , Humanos , Línea Celular Tumoral , Sulfotransferasas/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Sulfatos de Condroitina/metabolismo , Sulfatasas/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Movimiento Celular/efectos de los fármacos , Microambiente Tumoral , Proteoglicanos de Heparán Sulfato/metabolismo , Antígenos de Neoplasias , Biomarcadores de Tumor
3.
Nanoscale ; 16(24): 11538-11541, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38841880

RESUMEN

Aggregation-induced emission luminogens (AIEgens) enable highly sensitive and in situ visualization of sulfatase to benefit the early diagnosis of breast cancer (BC), but current sulfatase AIEgens always emit visible light (<650 nm). Herein, a near-infrared (NIR) AIEgen QMT-SFA is developed for sulfatase imaging in vivo. Hydrophilic QMT-SFA is cleaved by sulfatase to yield hydrophobic QMT-OH, which subsequently aggregates into nanoparticles to turn the AIE fluorescence "on", enabling sensitive sulfatase imaging in 4T1 cells and mouse models.


Asunto(s)
Neoplasias de la Mama , Sulfatasas , Animales , Femenino , Ratones , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Sulfatasas/metabolismo , Humanos , Colorantes Fluorescentes/química , Ratones Endogámicos BALB C , Nanopartículas/química , Rayos Infrarrojos , Ratones Desnudos
4.
Food Funct ; 15(10): 5439-5449, 2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38650575

RESUMEN

Barley, rich in bioactive components including dietary fiber, polyphenolic compounds and functional proteins, exhibits health benefits such as regulating glucose and lipid metabolism. Previous studies have found that the content and composition of free phenolic acids in barley may be significantly changed by fermentation with the laboratory patented strain Lactobacillus plantarum dy-1 (L. p dy-1), but the mechanism of enzymatic release of phenolic acid remains to be elucidated. Based on this, this study aimed to identify the key enzyme in L. p dy-1 responsible for releasing the bound phenolic acid and to further analyze its enzymatic properties. The Carbohydrate-Active enZYmes database revealed that L. p dy-1 encodes 7 types of auxiliary enzymes, among which we have identified a membrane sulfatase. The enzyme gene LPMS05445 was heterologous to that expressed in E. coli, and a recombinant strain was induced to produce the target protein and purified. The molecular weight of the purified enzyme was about 59.9 kDa, with 578.21 U mg-1 enzyme activity. The optimal temperature and pH for LPMS05445 expression were 40 °C and 7.0, respectively. Furthermore, enzymatic hydrolysis by LPMS05445 can obviously change the surface microstructure of dietary fiber from barley bran and enhance the release of bound phenolic acid, thereby increasing the free phenolic acid content and improving its physiological function. In conclusion, sulfatase produced by Lactobacillus plantarum dy-1 plays a key role in releasing bound phenolic acids during the fermentation of barley.


Asunto(s)
Lactobacillus plantarum , Sulfatasas , Lactobacillus plantarum/enzimología , Lactobacillus plantarum/metabolismo , Lactobacillus plantarum/genética , Sulfatasas/metabolismo , Sulfatasas/genética , Sulfatasas/química , Hordeum , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/química , Fermentación , Hidroxibenzoatos/metabolismo , Concentración de Iones de Hidrógeno , Escherichia coli/genética , Temperatura , Fibras de la Dieta/metabolismo
5.
Molecules ; 28(8)2023 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-37110530

RESUMEN

For nearly 30 years, resveratrol has attracted the scientific community's interest. This has happened thanks to the so-called French paradox, that is, the paradoxically low mortality from cardiovascular causes in the French population despite a diet rich in saturated fat. This phenomenon has been linked to the consumption of red wine, which contains a relatively high level of resveratrol. Currently, resveratrol is valued for its versatile, beneficial properties. Apart from its anti-atherosclerotic activity, resveratrol's antioxidant and antitumor properties deserve attention. It was shown that resveratrol inhibits tumour growth at all three stages: initiation, promotion, and progression. Moreover, resveratrol delays the ageing process and has anti-inflammatory, antiviral, antibacterial, and phytoestrogenic properties. These favorable biological properties have been demonstrated in vitro and in vivo in animal and human models. Since the beginning of the research on resveratrol, its low bioavailability, mainly due to its rapid metabolism, especially the first-pass effect that leaves almost no free resveratrol in the peripheral circulation, has been indicated as a drawback that has hindered its use. The elucidation of such issues as pharmacokinetics, stability, and the biological activity of resveratrol metabolites is therefore crucial for understanding the biological activity of resveratrol. Second-phase metabolism enzymes are mainly involved in RSV metabolism, e.g., UDP-glucuronyl transferases and sulfotransferases. In the present paper, we took a closer look at the available data on the activity of resveratrol sulfate metabolites and the role of sulfatases in releasing active resveratrol in target cells.


Asunto(s)
Estilbenos , Sulfotransferasas , Animales , Humanos , Resveratrol/farmacología , Sulfotransferasas/metabolismo , Sulfatasas/metabolismo , Antioxidantes/farmacología , Antioxidantes/metabolismo , Absorción Intestinal , Estilbenos/metabolismo
6.
J Cell Sci ; 136(7)2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36897575

RESUMEN

Morphogens provide quantitative and robust signaling systems to achieve stereotypic patterning and morphogenesis. Heparan sulfate (HS) proteoglycans (HSPGs) are key components of such regulatory feedback networks. In Drosophila, HSPGs serve as co-receptors for a number of morphogens, including Hedgehog (Hh), Wingless (Wg), Decapentaplegic (Dpp) and Unpaired (Upd, or Upd1). Recently, Windpipe (Wdp), a chondroitin sulfate (CS) proteoglycan (CSPG), was found to negatively regulate Upd and Hh signaling. However, the roles of Wdp, and CSPGs in general, in morphogen signaling networks are poorly understood. We found that Wdp is a major CSPG with 4-O-sulfated CS in Drosophila. Overexpression of wdp modulates Dpp and Wg signaling, showing that it is a general regulator of HS-dependent pathways. Although wdp mutant phenotypes are mild in the presence of morphogen signaling buffering systems, this mutant in the absence of Sulf1 or Dally, molecular hubs of the feedback networks, produces high levels of synthetic lethality and various severe morphological phenotypes. Our study indicates a close functional relationship between HS and CS, and identifies the CSPG Wdp as a novel component in morphogen feedback pathways.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Proteoglicanos Tipo Condroitín Sulfato/genética , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Proteoglicanos de Heparán Sulfato/genética , Proteoglicanos de Heparán Sulfato/metabolismo , Sulfatasas/genética , Sulfatasas/metabolismo , Proteína Wnt1/genética , Proteína Wnt1/metabolismo
7.
Org Lett ; 25(12): 2001-2005, 2023 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-36662590

RESUMEN

α-Formylglycine (fGly) is a rare residue located in the active site of sulfatases and serves as a precursor to pharmaceutically relevant motifs. The installation of fGly motifs into peptides is currently challenging due to degradation under the acidic and nucleophile-rich conditions accompanying resin cleavage during solid-phase peptide synthesis. We report the synthesis of acid- and nucleophile-tolerant α-formylglycine building blocks from vitamin C and use them to prepare callyaerin A, a macrocyclic peptide containing an fGly-derived motif.


Asunto(s)
Alanina , Técnicas de Síntesis en Fase Sólida , Alanina/química , Glicina/química , Sulfatasas/química , Sulfatasas/metabolismo , Péptidos/química
8.
Cell Mol Immunol ; 19(10): 1185-1195, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36068294

RESUMEN

Extracellular sulfatase-2 (Sulf-2) influences receptor-ligand binding and subsequent signaling by chemokines and growth factors, yet Sulf-2 remains unexplored in inflammatory cytokine signaling in the context of rheumatoid arthritis (RA). In the present study, we characterized Sulf-2 expression in RA and investigated its potential role in TNF-α-induced synovial inflammation using primary human RA synovial fibroblasts (RASFs). Sulf-2 expression was significantly higher in serum and synovial tissues from patients with RA and in synovium and serum from hTNFtg mice. RNA sequencing analysis of TNF-α-stimulated RASFs showed that Sulf-2 siRNA modulated ~2500 genes compared to scrambled siRNA. Ingenuity Pathway Analysis of RNA sequencing data identified Sulf-2 as a primary target in fibroblasts and macrophages in RA. Western blot, ELISA, and qRT‒PCR analyses confirmed that Sulf-2 knockdown reduced the TNF-α-induced expression of ICAM1, VCAM1, CAD11, PDPN, CCL5, CX3CL1, CXCL10, and CXCL11. Signaling studies identified the protein kinase C-delta (PKCδ) and c-Jun N-terminal kinase (JNK) pathways as key in the TNF-α-mediated induction of proteins related to cellular adhesion and invasion. Knockdown of Sulf-2 abrogated TNF-α-induced RASF proliferation. Sulf-2 knockdown with siRNA and inhibition by OKN-007 suppressed the TNF-α-induced phosphorylation of PKCδ and JNK, thereby suppressing the nuclear translocation and DNA binding activity of the transcription factors AP-1 and NF-κBp65 in human RASFs. Interestingly, Sulf-2 expression positively correlated with the expression of TNF receptor 1, and coimmunoprecipitation assays demonstrated the binding of these two proteins, suggesting they exhibit crosstalk in TNF-α signaling. This study identified a novel role of Sulf-2 in TNF-α signaling and the activation of RA synoviocytes, providing the rationale for evaluating the therapeutic targeting of Sulf-2 in preclinical models of RA.


Asunto(s)
Artritis Reumatoide , Sulfatasas/metabolismo , Factor de Necrosis Tumoral alfa , Animales , Artritis Reumatoide/metabolismo , Células Cultivadas , ADN/metabolismo , Fibroblastos/metabolismo , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ligandos , Ratones , Proteína Quinasa C-delta/metabolismo , ARN Interferente Pequeño/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Membrana Sinovial , Factor de Transcripción AP-1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
9.
Int J Mol Sci ; 23(15)2022 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-35897729

RESUMEN

Sulfatases are enzymes that catalyze the removal of sulfate from biological substances, an essential process for the homeostasis of the body. They are commonly activated by the unusual amino acid formylglycine, which is formed from cysteine at the catalytic center, mediated by a formylglycine-generating enzyme as a post-translational modification. Sulfatases are expressed in various cellular compartments such as the lysosome, the endoplasmic reticulum, and the Golgi apparatus. The substrates of mammalian sulfatases are sulfolipids, glycosaminoglycans, and steroid hormones. These enzymes maintain neuronal function in both the central and the peripheral nervous system, chondrogenesis and cartilage in the connective tissue, detoxification from xenobiotics and pharmacological compounds in the liver, steroid hormone inactivation in the placenta, and the proper regulation of skin humidification. Human sulfatases comprise 17 genes, 10 of which are involved in congenital disorders, including lysosomal storage disorders, while the function of the remaining seven is still unclear. As for the genes responsible for pathogenesis, therapeutic strategies have been developed. Enzyme replacement therapy with recombinant enzyme agents and gene therapy with therapeutic transgenes delivered by viral vectors are administered to patients. In this review, the biochemical substrates, disease manifestation, and therapy for sulfatases are summarized.


Asunto(s)
Enfermedades por Almacenamiento Lisosomal , Sulfatasas , Animales , Cisteína/metabolismo , Femenino , Humanos , Enfermedades por Almacenamiento Lisosomal/genética , Enfermedades por Almacenamiento Lisosomal/terapia , Mamíferos/metabolismo , Embarazo , Procesamiento Proteico-Postraduccional , Proteínas/metabolismo , Sulfatasas/genética , Sulfatasas/metabolismo
10.
Cancer Lett ; 538: 215693, 2022 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-35472437

RESUMEN

Owing to the lack of early diagnosis, pancreatic ductal adenocarcinoma (PDAC) remains one of the most lethal tumours. Because acinar-to-ductal metaplasia (ADM) is a critical process to pancreatic regeneration and PDAC initiation, we applied GSE65146, a dataset composed of transcripts at different time points in wild-type and KrasG12D mutant mice upon pancreatitis induction, to obtain regeneration- and tumour initiation-related genes. By overlapping with genes differentially expressed in human PDAC, we defined the initiation- and progression-related genes, and the most prognostic gene, SULF2, was selected for further verification. By using multiple PDAC genetically engineered murine models (GEMMs), we further verified that the expression of SULF2 was increased at the ADM and PDAC stages. Functionally, SULF2 was able to promote the dedifferentiation of acinar cells as well as the metastatic ability of PDAC. Additionally, our study revealed that SULF2 could enhance TGFß-SMAD signalling via GDF15. More importantly, serum SULF2 was elevated in patients with PDAC, and in combination with CA19-9, it provided a better method for PDAC diagnosis. Herein, our study screened out key genes for the initiation and progression of PDAC, providing potential indicators for the diagnosis of the disease.


Asunto(s)
Carcinoma Ductal Pancreático , Factor 15 de Diferenciación de Crecimiento , Neoplasias Pancreáticas , Proteínas Smad , Sulfatasas , Células Acinares , Animales , Carcinoma Ductal Pancreático/patología , Progresión de la Enfermedad , Factor 15 de Diferenciación de Crecimiento/genética , Factor 15 de Diferenciación de Crecimiento/metabolismo , Humanos , Ratones , Neoplasias Pancreáticas/patología , Sulfatasas/metabolismo
11.
Cell Rep ; 38(11): 110516, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35294879

RESUMEN

Sulfs represent a class of unconventional sulfatases which provide an original post-synthetic regulatory mechanism for heparan sulfate polysaccharides and are involved in multiple physiopathological processes, including cancer. However, Sulfs remain poorly characterized enzymes, with major discrepancies regarding their in vivo functions. Here we show that human Sulf-2 (HSulf-2) harbors a chondroitin/dermatan sulfate glycosaminoglycan (GAG) chain, attached to the enzyme substrate-binding domain. We demonstrate that this GAG chain affects enzyme/substrate recognition and tunes HSulf-2 activity in vitro and in vivo. In addition, we show that mammalian hyaluronidase acts as a promoter of HSulf-2 activity by digesting its GAG chain. In conclusion, our results highlight HSulf-2 as a proteoglycan-related enzyme and its GAG chain as a critical non-catalytic modulator of the enzyme activity. These findings contribute to clarifying the conflicting data on the activities of the Sulfs.


Asunto(s)
Dermatán Sulfato , Sulfotransferasas , Animales , Heparitina Sulfato , Humanos , Mamíferos/metabolismo , Unión Proteica , Sulfatasas/metabolismo , Sulfotransferasas/genética , Sulfotransferasas/metabolismo
12.
Methods Mol Biol ; 2303: 415-425, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34626397

RESUMEN

Extracellular sulfatases (SULF1 and SULF2) selectively remove 6-O-sulfate groups (6OS) from heparan sulfate proteoglycans (HSPGs) and by this process control important interactions of HSPGs with extracellular factors including morphogens, growth factors, and extracellular matrix (ECM) components. The expression of SULF1 and SULF2 is dynamically regulated during development and is altered in pathological states such as glioblastoma (GBM), a highly malignant and highly invasive brain cancer. SULF2 protein is increased in an important subset of human GBM and it helps regulate receptor tyrosine kinase (RTK) signaling and tumor growth in a murine model of the disease. By altering ligand binding to HSPGs SULF2 has the potential to modify the extracellular availability of factors important in a number of cell processes including proliferation, chemotaxis, and migration. Diffuse invasion of malignant tumor cells into surrounding healthy brain is a characteristic feature of GBM that makes therapy challenging. Here, we describe methods to assess SULF2 expression in human tumor tissue and cell lines and how to relate this to tumor cell invasion.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Animales , Humanos , Ratones , Transducción de Señal , Sulfatasas/genética , Sulfatasas/metabolismo , Sulfotransferasas/genética , Sulfotransferasas/metabolismo
13.
Sci Rep ; 11(1): 22424, 2021 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-34789772

RESUMEN

Sulf1/Sulf2 genes are highly expressed during early fetal cardiovascular development but down-regulated during later stages correlating with a number of cell signalling pathways in a positive or a negative manner. Immunocytochemical analysis confirmed SULF1/SULF2 expression not only in endothelial cell lining of blood vessels but also in the developing cardiomyocytes but not in the adult cardiomyocytes despite persisting at reduced levels in the adult endothelial cells. The levels of both SULFs in adult ischemic human hearts and in murine hearts following coronary occlusion increased in endothelial lining of some regional blood vessels but with little or no detection in the cardiomyocytes. Unlike the normal adult heart, the levels of SULF1 and SULF2 were markedly increased in the adult canine right-atrial haemangiosarcoma correlating with increased TGFß cell signalling. Cell signalling relationship to ischaemia was further confirmed by in vitro hypoxia of HMec1 endothelial cells demonstrating dynamic changes in not only vegf and its receptors but also sulfotransferases and Sulf1 & Sulf2 levels. In vitro hypoxia of HMec1 cells also confirmed earlier up-regulation of TGFß cell signalling revealed by Smad2, Smad3, ALK5 and TGFß1 changes and later down-regulation correlating with Sulf1 but not Sulf2 highlighting Sulf1/Sulf2 differences in endothelial cells under hypoxia.


Asunto(s)
Corazón/embriología , Infarto del Miocardio/metabolismo , Transducción de Señal , Sulfatasas/metabolismo , Sulfotransferasas/metabolismo , Animales , Hipoxia de la Célula , Línea Celular , Modelos Animales de Enfermedad , Perros , Células Endoteliales/metabolismo , Humanos , Ratones , Infarto del Miocardio/patología , Miocardio/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
14.
Angew Chem Int Ed Engl ; 60(36): 19957-19964, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34164914

RESUMEN

Aminomalonate (Ama) is a widespread structural motif in Nature, whereas its biosynthetic route is only partially understood. In this study, we show that a radical S-adenosylmethionine (rSAM) enzyme involved in cyclophane biosynthesis exhibits remarkable catalytic promiscuity. This enzyme, named three-residue cyclophane forming enzyme (3-CyFE), mainly produces cyclophane in vivo, whereas it produces formylglycine (FGly) as a major product and barely produce cyclophane in vitro. Importantly, the enzyme can further oxidize FGly to produce Ama. Bioinformatic study revealed that 3-CyFEs have evolved from a common ancestor with anaerobic sulfatase maturases (anSMEs), and possess a similar set of catalytic residues with anSMEs. Remarkably, the enzyme does not need leader peptide for activity and is fully active on a truncated peptide containing only 5 amino acids of the core sequence. Our work discloses the first ribosomal path towards Ama formation, providing a possible hint for the rich occurrence of Ama in Nature.


Asunto(s)
Malonatos/metabolismo , Péptidos/metabolismo , S-Adenosilmetionina/metabolismo , Sulfatasas/metabolismo , Radicales Libres/química , Radicales Libres/metabolismo , Malonatos/química , Estructura Molecular , Péptidos/química , Procesamiento Proteico-Postraduccional , S-Adenosilmetionina/química , Sulfatasas/química
15.
Cells ; 10(4)2021 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-33923753

RESUMEN

During angiogenesis, vascular endothelial growth factor A (VEGFA) regulates endothelial cell (EC) survival, tip cell formation, and stalk cell proliferation via VEGF receptor 2 (VEGFR2). VEGFR2 can interact with VEGFR2 co-receptors such as heparan sulfate proteoglycans (HSPGs) and neuropilin 2 (NRP2), but the exact roles of these co-receptors, or of sulfatase 2 (SULF2), an enzyme that removes sulfate groups from HSPGs and inhibits HSPG-mediated uptake of very low density lipoprotein (VLDL), in angiogenesis and tip cell biology are unknown. In the present study, we investigated whether the modulation of binding of VEGFA to VEGFR2 by knockdown of SULF2 or NRP2 affects sprouting angiogenesis, tip cell formation, proliferation of non-tip cells, and EC survival, or uptake of VLDL. To this end, we employed VEGFA splice variant 121, which lacks an HSPG binding domain, and VEGFA splice variant 165, which does have this domain, in in vitro models of angiogenic tip cells and vascular sprouting. We conclude that VEGFA165 and VEGFA121 have similar inducing effects on tip cells and sprouting in vitro, and that the binding of VEGFA165 to HSPGs in the extracellular matrix does not seem to play a role, as knockdown of SULF2 did not alter these effects. Co-binding of NRP2 appears to regulate VEGFA-VEGFR2-induced sprout initiation, but not tip cell formation. Finally, as the addition of VLDL increased sprout formation but not tip cell formation, and as VLDL uptake was limited to non-tip cells, our findings suggest that VLDL plays a role in sprout formation by providing biomass for stalk cell proliferation.


Asunto(s)
Heparitina Sulfato/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Neovascularización Fisiológica , Neuropilina-2/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Apoptosis , Humanos , Lipoproteínas VLDL/metabolismo , Sulfatasas/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
16.
Hepatology ; 74(3): 1411-1428, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33735525

RESUMEN

BACKGROUND AND AIMS: Existing therapeutic approaches to treat cholangiocarcinoma (CCA) have limited effectiveness, prompting further study to develop therapies for CCA. We report a mechanistic role for the heparan sulfate editing enzyme sulfatase 2 (SULF2) in CCA pathogenesis. APPROACH AND RESULTS: In silico analysis revealed elevated SULF2 expression in human CCA samples, occurring partly through gain of SULF2 copy number. We examined the effects of knockdown or overexpression of SULF2 on tumor growth, chemoresistance, and signaling pathway activity in human CCA cell lines in vitro. Up-regulation of SULF2 in CCA leads to increased platelet-derived growth factor receptor beta (PDGFRß)-Yes-associated protein (YAP) signaling activity, promoting tumor growth and chemotherapy resistance. To explore the utility of targeting SULF2 in the tumor microenvironment for CCA treatment, we tested an anti-SULF2 mouse monoclonal antibody, 5D5, in a mouse CCA xenograft model. Targeting SULF2 by monoclonal antibody 5D5 inhibited PDGFRß-YAP signaling and tumor growth in the mouse xenograft model. CONCLUSIONS: These results suggest that SULF2 monoclonal antibody 5D5 or related agents may be potentially promising therapeutic agents in CCA.


Asunto(s)
Neoplasias de los Conductos Biliares/genética , Colangiocarcinoma/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Sulfatasas/genética , Proteínas Señalizadoras YAP/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Neoplasias de los Conductos Biliares/metabolismo , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Colangiocarcinoma/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Trasplante de Neoplasias , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/efectos de los fármacos , Sulfatasas/antagonistas & inhibidores , Sulfatasas/metabolismo , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas Señalizadoras YAP/efectos de los fármacos
17.
Mini Rev Med Chem ; 21(20): 3144-3165, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33463462

RESUMEN

Breast cancer is the most frequently diagnosed cancer in women and the second most common form of cancer, causing death after lung cancer, all across the globe at an alarming rate. The level of estrogens in breast cancer tissues of postmenopausal women is 10-40 folds higher than the non-carcinogenic breast tissues. As a result of this greater level of estrogen, breast tissue becomes more prone to develop breast cancer; mainly, estradiol plays a significant role in the initiation and development of hormone-dependent breast cancer. Androstenedione, Adrenal dehydroepiandrosterone sulfate, and estrone-sulfate also play an important role as precursors for estrogen biosynthesis. Estrogen deprivation exhibits an attractive phenomenon in the advancement of ideal therapeutics for the treatment of breast cancer. Inhibition of aromatase and sulphatase emerged as an attractive therapy for the treatment of hormone-dependent breast cancer via deprivation of estrogen by different pathways. The cocktail of aromatase and sulphatase inhibitors known as Dual Aromatase-sulphatase Inhibitors (DASIs) emerged as an attractive approach for effective estrogen deprivation. The present review article focused on the journey of dual aromatase-sulphatase inhibitors from the beginning to date (2020). Keeping in view the key observations, this review may be helpful for medicinal chemists to design and develop new and efficient dual aromatase-sulphatase inhibitors for the possible treatment of hormone- related breast cancer.


Asunto(s)
Inhibidores de la Aromatasa , Neoplasias de la Mama , Aromatasa/metabolismo , Inhibidores de la Aromatasa/farmacología , Inhibidores de la Aromatasa/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Estrógenos , Femenino , Humanos , Sulfatasas/metabolismo
18.
Eur J Med Chem ; 213: 113037, 2021 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-33257172

RESUMEN

Cancer is the world's devastating disease, and breast cancer is the most common reason for the death of women worldwide. Many synthetic drugs and medications are provided with their beneficial actions, but all of these have side effects and resistance problems. Natural remedies are coming forward to overcome the disadvantages of synthetic drugs. Among the natural categories, phytoestrogens having a structural similarity of mammalian oestradiol proves its benefit with various mechanisms not only in the treatment of breast cancer but even to prevent the occurrence of postmenopausal symptoms. Phytoestrogens are plant-derived compounds that were utilized in ancient medications and traditional knowledge for its sex hormone properties. Phytoestrogens exert pleiotropic effects on cellular signalling and show effects on estrogen-dependent diseases. However, because of activation/inhibition of steroid hormonal receptor ER-α or ER-ß, these compounds induce or inhibit steroid hormonal (estrogen) action and, therefore, have the potential to disrupt hormone (estrogen) signalling pathway. In this review, we have discussed and summarize the effect of certain phytoestrogens and their possible mechanisms that can substantiate advantageous benefits for the treatment of post-menopausal symptoms as well as for breast cancer.


Asunto(s)
Antineoplásicos Fitogénicos/química , Neoplasias de la Mama/tratamiento farmacológico , Fitoestrógenos/química , Animales , Antineoplásicos Fitogénicos/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Estradiol/química , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Estrógenos/metabolismo , Femenino , Flavonoides/química , Humanos , Lignanos/química , Fitoestrógenos/farmacología , Transducción de Señal , Estilbenos/química , Relación Estructura-Actividad , Sulfatasas/metabolismo
19.
Int J Mol Sci ; 21(20)2020 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-33076548

RESUMEN

The natural flavonoid chrysin possesses antiproliferative activity against various types of cancers, including hepatocellular carcinoma (HCC), which is a common malignancy. However, the exact mechanism of chrysin antiproliferative activity remains unclear. This research was executed to explore the impact of chrysin on glypican-3 (GPC3)/sulfatase-2 (SULF2) axis and lncRNA-AF085935 expression in HCC using HepG2 cells. Cisplatin (20, 50, 100 µg/mL), chrysin (15, 30, and 60 µg/mL) and the combination of 50 µg/mL cisplatin with different concentrations of chrysin were applied for 24/48 h. Cell viability was determined by MTT assay. Protein levels of GPC3 and SULF2 were measured by ELISA at 24/48 h. GPC3 immunoreactivity was detected by immunocytochemistry. Moreover, GPC3 and SULF2 mRNA expressions in addition to lncRNA-AF085935 expression were assessed by qPCR at 48 h. The GPC3 protein, immunostaining and mRNA levels, SULF2 protein and mRNA levels, as well as lncRNA-AF085935 expression, were decreased significantly with cisplatin and chrysin alone when compared with the control untreated HepG2 cells. However, the combination treatment exhibited a better chemopreventive effect in a dose- and time-dependent manner. This study demonstrated, for the first time, the antiproliferative activity of chrysin against HCC through the suppression of the GPC3/SULF2 axis along with the downregulation of lncRNA-AF085935 expression. Synergistic effect of chrysin with cisplatin could potentiate their antiproliferative action in a dose- and time-dependent manner.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Hepatocelular/metabolismo , Flavonoides/farmacología , Neoplasias Hepáticas/metabolismo , ARN Largo no Codificante/metabolismo , Proliferación Celular/efectos de los fármacos , Glipicanos/genética , Glipicanos/metabolismo , Células Hep G2 , Humanos , ARN Largo no Codificante/genética , Sulfatasas/genética , Sulfatasas/metabolismo
20.
Biochem J ; 477(20): 3963-3983, 2020 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-33120425

RESUMEN

Sulfatases constitute a family of enzymes that specifically act in the hydrolytic degradation of sulfated metabolites by removing sulfate monoesters from various substrates, particularly glycolipids and glycosaminoglycans. A common essential feature of all known eukaryotic sulfatases is the posttranslational modification of a critical cysteine residue in their active site by oxidation to formylglycine (FGly), which is mediated by the FGly-generating enzyme in the endoplasmic reticulum and is indispensable for catalytic activity. The majority of the so far described sulfatases localize intracellularly to lysosomes, where they act in different catabolic pathways. Mutations in genes coding for lysosomal sulfatases lead to an accumulation of the sulfated substrates in lysosomes, resulting in impaired cellular function and multisystemic disorders presenting as lysosomal storage diseases, which also cover the mucopolysaccharidoses and metachromatic leukodystrophy. Bioinformatics analysis of the eukaryotic genomes revealed, besides the well described and long known disease-associated sulfatases, additional genes coding for putative enzymes with sulfatases activity, including arylsulfatase G as well as the arylsulfatases H, I, J and K, respectively. In this article, we review current knowledge about lysosomal sulfatases with a special focus on the just recently characterized family members arylsulfatase G and arylsulfatase K.


Asunto(s)
Enfermedades por Almacenamiento Lisosomal/enzimología , Lisosomas/metabolismo , Sulfatasas/genética , Sulfatasas/metabolismo , Animales , Dominio Catalítico , Modelos Animales de Enfermedad , Retículo Endoplásmico/metabolismo , Glicina/análogos & derivados , Glicina/química , Humanos , Lisosomas/enzimología , Filogenia , Procesamiento Proteico-Postraduccional , Sulfatasas/química , Sulfatasas/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA