Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Br J Pharmacol ; 181(16): 2774-2793, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38644540

RESUMEN

BACKGROUND AND PURPOSE: White adipose tissue (WAT) is involved in rheumatoid arthritis (RA). This study explored its potential as an antirheumatic target. EXPERIMENTAL APPROACH: WAT status of healthy and adjuvant-induced arthritis (AIA) rats were compared. The contribution of WAT to RA pathology was evaluated by pre-adipocyte transplant experiments and by dissecting perirenal fat pads of AIA rats. The impact of RA on WAT was investigated by culturing pre-adipocytes. Proteins differentially expressed in WAT of healthy and AIA rats were identified by the UPLC/MS2 method. These together with PPARγ siRNA and agonist were used to treat pre-adipocytes in vitro. The medium was used for THP-1 monocyte culture. KEY RESULTS: Compared with healthy controls, AIA WAT was smaller but secreted more leptin, eNAMPT, MCP-1, TNF-α, and IL-6. AIA rat pre-adipocytes increased the levels of these adipokines in healthy recipients. RA patients' serum induced a similar secretion change and impaired differentiation of pre-adipocytes. Adipectomy eased AIA-related immune abnormalities and arthritic manifestations. Hepatokines PON1, IGFBP4, and GPIHBP1 were among the differential proteins in high levels in RA blood, and induced inflammatory secretions by pre-adipocytes. GPIHBP1 inhibited PPARγ expression and caused differentiation impairment and inflammatory secretion by pre-adipocytes, a similar outcome to PPARγ-silencing. This endowed the cells with an ability to activate monocytes, which can be abrogated by rosiglitazone. CONCLUSION AND IMPLICATIONS: Certain hepatokines potentiate inflammatory secretions by pre-adipocytes and expedite RA progression by inhibiting PPARγ. Targeting this signalling or abnormal WAT secretion by various approaches may reduce RA severity.


Asunto(s)
Tejido Adiposo Blanco , Artritis Experimental , Artritis Reumatoide , PPAR gamma , Animales , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo Blanco/efectos de los fármacos , Humanos , Ratas , Artritis Experimental/metabolismo , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/patología , Masculino , Artritis Reumatoide/metabolismo , Artritis Reumatoide/tratamiento farmacológico , PPAR gamma/metabolismo , PPAR gamma/agonistas , Antirreumáticos/farmacología , Antirreumáticos/uso terapéutico , Femenino , Ratas Endogámicas Lew , Adipocitos/metabolismo , Adipocitos/efectos de los fármacos , Adipoquinas/metabolismo
2.
Drug Des Devel Ther ; 17: 563-577, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36860800

RESUMEN

Background: α-Mangostin (MG) showed the potentials in alleviating experimental arthritis, inhibiting inflammatory polarization of macrophages/monocytes, and regulating peroxisome proliferators-activated receptor γ (PPAR-γ) and silent information regulator 1 (SIRT1) signals. The aim of this study was to analyze the correlations among the above-mentioned properties. Methods: Antigen-induced arthritis (AIA) was established in mouse, which was treated with MG in combination with SIRT1/PPAR-γ inhibitors to clarify the role of the two signals in the anti-arthritic actions. Pathological changes were systematically investigated. Phenotypes of cells were investigated by flow cytometry. Expression and co-localization of SIRT1 and PPAR-γ proteins in joint tissues were observed by the immunofluorescence method. Finally, clinical implications from the synchronous up-regulation of SIRT1 and PPAR-γ were validated by experiments in vitro. Results: SIRT1 and PPAR-γ inhibitors (nicotinamide and T0070097) reduced the therapeutic effects of MG on AIA mice, and abrogated MG-induced up-regulation of SIRT1/PPAR-γ and inhibition of M1 polarization in macrophages/monocytes. MG has a good binding affinity to PPAR-γ, and MG promoted the co-expression of SIRT1 and PPAR-γ in joints. Synchronously activating SIRT1 and PPAR-γ was revealed to be necessary by MG to repress inflammatory responses in THP-1 monocytes. Conclusion: MG binds PPAR-γ and excites this signaling to initiate ligand-dependent anti-inflammatory activity. Due to certain unspecified signal transduction crosstalk mechanism, it then promoted SIRT1 expression and further limited inflammatory polarization of macrophages/monocytes in AIA mice.


Asunto(s)
Artritis Experimental , Monocitos , Animales , Ratones , Proliferadores de Peroxisomas , PPAR gamma , Sirtuina 1 , Macrófagos , Artritis Experimental/inducido químicamente , Artritis Experimental/tratamiento farmacológico
3.
Immunopharmacol Immunotoxicol ; 45(1): 16-25, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35850595

RESUMEN

BACKGROUND: The first-line anti-rheumatic drug methotrexate (MTX) is used in the combination. Because of the unpredictable adverse reactions, optimization of relevant regimens is necessary and meaningful. This study aimed to study the possible interaction between Securidaca inappendiculate Hassk. Derived xanthones and MTX. METHODS: We established adjuvant-induced arthritis (AIA) model, which was treated with MTX and MTX + xanthone-rich fraction (XRF). The clinical efficacy was evaluated by histopathological examination, and LC-MS was used to monitor the blood concentration of MTX. Western blotting and immunohistochemistry were used to detect protein expression. In vitro, we assessed the activity of related transporters by cellular uptake assay based on HEK-293T cells. RESULTS: Compared with MTX-treated rats, inflammation in the immunized rats in the MTX + XRF group was obvious, indicating that XRF antagonized the anti-rheumatic effect of MTX. Meanwhile, XRF reduced liver and kidney injuries caused by MTX in addition to MTX. Results from immunohistochemical and nappendiculat assays suggested that XRF may reduce uptake of MTX by down-regulating reduced folate carrier 1 (RFC1). CONCLUSION: This study indicated that XRF could reduce the plasma concentration of MTX by inhibiting the expression of RFC1, antagonize the therapeutic effect of MTX on AIA rats, and reduce its oral bioavailability. The combination of S. inappendiculate and MTX should be further optimized to achieve the goal of increasing efficiency and reducing toxicity.


Asunto(s)
Antirreumáticos , Securidaca , Xantonas , Ratas , Animales , Metotrexato/farmacología , Securidaca/metabolismo , Proteína Portadora de Folato Reducido , Antirreumáticos/farmacología , Antirreumáticos/uso terapéutico , Xantonas/farmacología
4.
Biomed Pharmacother ; 150: 113067, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35658235

RESUMEN

Methotrexate (MTX) is a first line anti-rheumatic drug. This study was designed to investigate the impact of rheumatoid arthritis (RA) conditions on its oral absorption, and clarify the relevance with changes of MTX absorption-related transporters in rheumatic models. MTX was orally administered to healthy, collagen-induced arthritis (CIA), and adjuvant-induced arthritis (AIA) rats. MTX plasma concentrations were determined by a validated liquid chromatography-mass spectrometry method. We found that intestinal MTX absorption was significantly increased in CIA/AIA rats versus healthy controls. This finding was supported by small intestine-based MTX uptake assay in vitro. Meanwhile, intestinal expression of both reduced folate carrier 1 (RCF1) and proton-coupled folate transporter (PCFT) remained unchanged. The everted intestinal sac assay confirms RFC1 is the key transporter accounting for intestinal MTX absorption, as its antagonist salicylazosulfapyridine showed potent capacity in reducing MTX uptake. No correlation between RA-related cytokines and RCF1 expression was observed in clinical samples. We further revealed that when cultured with AIA rat or RA patient serum, lactate and adenosine triphosphate (ATP) production as well as MTX uptake in MDCKII cells were significantly increased, and this increase was completely abrogated by ATP production-related metabolic inhibitors. Thanks to its inhibitory effects on MTX bioavailability, the glycolysis inhibitor shikonin diminished MTX-induced injuries of kidney and liver in AIA rats. These data demonstrate that glycolysis-driven high energy metabolism increases MTX absorption in rheumatic subjects, leading to the exacerbated toxicity. These findings will have important implications in optimizing MTX regimens for RA treatment with better efficacy and lower toxicity.


Asunto(s)
Antirreumáticos , Artritis Experimental , Artritis Reumatoide , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Adenosina Trifosfato/metabolismo , Animales , Antirreumáticos/uso terapéutico , Antirreumáticos/toxicidad , Artritis Experimental/tratamiento farmacológico , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/metabolismo , Glucólisis , Humanos , Absorción Intestinal , Metotrexato/farmacología , Ratas
5.
Int Immunopharmacol ; 98: 107856, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34130151

RESUMEN

Supplement of nicotinamide mononucleotide (NMN), the direct precursor of nicotinamide adenine dinucleotide (NAD+) has gained prominence due to the significant anti-aging potentials of nicotinamide phosphoribosyltransferas (NAMPT)/NAD+ signaling. Because over-expression of NAMPT is deeply implicated in inflammatory arthritis, we investigated the effects of NMN supplement on rats with adjuvant-induced arthritis (AIA). Tested rats were given oral treatment of NMN at 200 mg/kg/day for 25 days. Arthritis score and body weight were periodically recorded. Clinical outcomes were evaluated based on arthritic manifestations, ELISA analysis and histological examination. T cells subsets were analyzed by flow cytometry. Expressions of protein and mRNA were assessed by immunoblotting and PCR methods, respectively. Levels of CD172a, CD43, and NAMPT in peripheral blood mononuclear cells (PBMCs) were investigated by immunofluorescence approach. Obtained results were further validated by experiments in vitro. Generally, NMN exacerbated AIA severity in rats. It deteriorated MMP3-controlled tissues damages, and altered immune profile by increasing Th17/Treg cells ratio. The up-regulation of NAMPT in PBMCs from NMN-treated rats was confirmed by both immunofluorescence and PCR experiments, which was synchronized with significant increase in iNOS, MCP-1, IL-1ß expression. NMN-primed AIA PBMCs were potent in up-regulating MCP-1, IL-1ß, MMP3 and p-JNK expression in synovioblast. NMN stimulus barely affected Th17 cells count in in vitro cultured splenocytes, but it greatly potentiated the capability of AIA monocytes in inducing IL-17α secretion and Th17 cells differentiation in the co-cultured splenocytes. It suggested that long-term NMN supplement could exacerbate inflammatory arthritis by reshaping the immune milieu through the up-regulation of NAMPT.


Asunto(s)
Artritis Experimental/inmunología , Artritis Reumatoide/inmunología , Citocinas/metabolismo , Nicotinamida Fosforribosiltransferasa/metabolismo , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Animales , Diferenciación Celular , Humanos , Inmunidad Colectiva , Masculino , Mononucleótido de Nicotinamida/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal
6.
Plant Physiol Biochem ; 70: 1-6, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23770588

RESUMEN

Untreated and Se-enriched apple leaves (Malus domestica Borkh. cv. 'Red Fuji') were used as the experimental materials. Proteomes of the differentially prepared tissues were compared through two-dimensional electrophoresis analysis and mass spectrum identification. There were 505 more protein spots in the proteome of the Se-enriched leaves than in the control leaves. Forty-seven protein spots were significantly differentially expressed (P < 0.05), among those, 32 protein spots were up-regulated while 12 protein points were down-regulated, and three new protein spots were found with the relative molecular masses of 31, 29, 26 kDa. Twenty-three protein spots with good shape and significant expression were selected for mass spectrometry analysis. These spots were excised from the gel and analyzed by a matrix-assisted laser desorption ionization time of flight mass spectrometry (MALDI-TOF-MS). Peptide mass fingerprints (PMF) of all the proteins were submitted to NCBInr for protein identification, and 10 differential proteins were positively identified. Biological information of the identified proteins was found via http://www.uniprot.org/. There were three (1475, 1479, 1527) ribulose-1,5-bisphosphate carboxylase/oxygenase large subunits (Rubisco), two ribulose-1,5-bisphosphate carboxylases (346, 486) belonging to the Rubisco large chain family, one photosystem I reaction center subunit II (297), one chloroplast oxygen-evolving enhancer protein 1 (619), one Os12g0127100 protein whose function was unknown (927), one monodehydroascorbate reductase (1451), and one polyphenol oxidase V (1596). The major subcellular location for these proteins was the chloroplast, and they play important roles in photosynthesis and stress resistance for plants.


Asunto(s)
Malus/metabolismo , Fotosíntesis , Hojas de la Planta/metabolismo , Proteínas de Plantas/metabolismo , Proteoma/metabolismo , Selenio/metabolismo , Adaptación Fisiológica , Cloroplastos/metabolismo , Malus/efectos de los fármacos , Malus/fisiología , Fotosíntesis/efectos de los fármacos , Hojas de la Planta/efectos de los fármacos , Hojas de la Planta/fisiología , Proteómica/métodos , Selenio/farmacología
7.
PLoS One ; 6(11): e26939, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22132082

RESUMEN

A new psychrophilic marine protease was found from a marine bacterium Flavobacterium YS-80 in the Chinese Yellow Sea. The protease is about 49 kD with an isoelectric point about 4.5. It consists of 480 amino acids and is homologous to a psychrophilic alkaline protease (PAP) from an Antarctic Pseudomonas species. The protein was purified from the natural bacterium fermented and crystallized. Its crystal structure (PDB ID 3U1R) was solved at 2.0 Å by Molecular Replacement using a model based on PAP, and was refined to a crystallographic R(work) of 0.16 and an R(free) of 0.21. The marine protease consists of a two domain structure with an N-terminal domain including residues 37-264 and a C-terminal domain including residues 265-480. Similar to PAP, the N-terminal domain is responsible for proteolysis and the C-terminal is for stability. His186, His190, His196 and Tyr226 are ligands for the Zn(2+) ion in the catalytic center. The enzyme's Tyr226 is closer to the Zn(2+) ion than in PAP and it shows a stronger Zn(2+)-Tyr-OH bond. There are eight calcium ions in the marine protease molecule and they have significantly shorter bond distances to their ligands compared to their counterparts in all three crystal forms of PAP. On the other hand, the loops in the marine protease are more compact than in PAP. This makes the total structure stable and less flexible, resulting in higher thermo stability. These properties are consistent with the respective environments of the proteases. The structural analysis of this new marine protease provides new information for the study of psychrophilic proteases and is helpful for elucidating the structure-environment adaptation of these enzymes.


Asunto(s)
Organismos Acuáticos/enzimología , Flavobacterium/enzimología , Metaloendopeptidasas/química , Secuencia de Aminoácidos , Calcio/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Iones , Ligandos , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Estructura Secundaria de Proteína , Alineación de Secuencia , Espectrofotometría Atómica , Zinc/metabolismo
8.
Fitoterapia ; 82(4): 626-31, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21300141

RESUMEN

BACKGROUND AND AIM: Hyperin, a flavonol compound extracted from the Chinese herb Abelmoschus manihot L. Medic, is reported to exert protective actions in cerebral ischemic injury. The specific aim of the present study was to study the relaxation of Hyperin in rat isolated basilar artery and identify the underlying cellular mechanisms. METHODS: Rat isolated basilar artery segments were cannulated and perfused while being superfused with PSS solution. Vessel images were recorded by video microscopy and diameters measured. Membrane potential was recorded using glass microelectrodes to evaluate the basilar artery smooth muscle cell hyperpolarization. RESULTS: Perfusion of Hyperin (1~100 µM) elicited a concentration-dependent relaxation of basilar artery segments preconstricted with 0.1 µM U46619. The response was significantly inhibited by the removal of the endothelium. Hyperin also elicited marked and concentration-dependent hyperpolarization of smooth muscle cells. 30 µM nitro-L-arginine (an inhibitor of nitric oxide synthase) and indomethacin (an inhibitor of cyclooxygenase), partially inhibited Hyperin-induced relaxation and hyperpolarization leaving an attenuated, but significant, endothelium-dependent relaxation and hyperpolarization. This remaining effect was almost completely blocked by 1mM tetraethylammonium (an inhibitor of Ca(2+)-activated K(+) channels), or by 100 µM DL-propargylglycine, an inhibitor of cystathionine-γ-lyase (a synthase of the endogenous H(2)S). CONCLUSION: These findings show that Hyperin produces significant hyperpolarization in rat basilar artery smooth muscle cells and relaxation through both endothelium-dependent and endothelium-independent mechanisms. The underlying mechanisms appeared to be multi-factorial involving nitric oxide, prostacyclin, and endothelium-derived hyperpolarizing factor (EDHF). Our data further suggest that endogenous H(2)S is a component of the EDHF-mediated hyperpolarization and relaxation to Hyperin.


Asunto(s)
Arteria Basilar/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Sulfuro de Hidrógeno/metabolismo , Quercetina/análogos & derivados , Vasodilatación/efectos de los fármacos , Alquinos , Animales , Medicamentos Herbarios Chinos/farmacología , Endotelio Vascular/metabolismo , Glicina/análogos & derivados , Indometacina , Masculino , Potenciales de la Membrana/efectos de los fármacos , Nitroarginina , Quercetina/farmacología , Ratas , Ratas Sprague-Dawley , Tetraetilamonio
9.
Toxicon ; 54(6): 709-16, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19500614

RESUMEN

Phospholipases A(2) (PLA(2)s) from snake venoms have diverse pharmacological functions including neurotoxicity, and more studies are necessary to understand relevant mechanisms. Here we report the different crystal structures for two enzymatically active basic subunits (HDP-1P and HDP-2P) of heterodimeric neurotoxic PLA(2)s isolated from Vipera nikolskii venom. Structural comparisons with similar PLA(2)s clearly show some flexible regions which might be important for the catalytic function and neurotoxicity. Unexpectedly, Triton X-100 molecule bound in the hydrophobic channel of HDP-1P and HDP-2P was observed, and its binding induced conformational changes in the Ca(2+) binding loop. Enzymatic activity measurements indicated that Triton X-100 decreased the activity of PLA(2), although with comparatively low inhibitory activity. For the first time exocytosis experiments in pancreatic beta cells were used to confirm the presynaptic neurotoxicity of relevant snake PLA(2). These experiments also indicated that Triton X-100 inhibited the influence of HDP-1P on exocytosis, but the inhibition was smaller than that of MJ33, a phospholipid-analogue inhibitor of PLA(2). Our studies performed at a cellular level are in good agreement with earlier findings that enzymatic activity of the snake presynaptic PLA(2) neurotoxins is essential for effective block of nerve terminals.


Asunto(s)
Octoxinol/química , Fosfolipasas A2/química , Fosfolipasas A2/toxicidad , Venenos de Víboras/enzimología , Secuencia de Aminoácidos , Animales , Dominio Catalítico , Línea Celular , Cristalización , Dimerización , Modelos Moleculares , Datos de Secuencia Molecular , Fosfolipasas A2/aislamiento & purificación , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad
10.
J Mol Biol ; 366(5): 1603-14, 2007 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-17224163

RESUMEN

Phosphoribosylaminoimidazole carboxylase/phosphoribosylaminoimidazole succinocarboxamide synthetase (PAICS) is an important bifunctional enzyme in de novo purine biosynthesis in vertebrate with both 5-aminoimidazole ribonucleotide carboxylase (AIRc) and 4-(N-succinylcarboxamide)-5-aminoimidazole ribonucleotide synthetase (SAICARs) activities. It becomes an attractive target for rational anticancer drug design, since rapidly dividing cancer cells rely heavily on the purine de novo pathway for synthesis of adenine and guanine, whereas normal cells favor the salvage pathway. Here, we report the crystal structure of human PAICS, the first in the entire PAICS family, at 2.8 A resolution. It revealed that eight PAICS subunits, each composed of distinct AIRc and SAICARs domains, assemble a compact homo-octamer with an octameric-carboxylase core and four symmetric periphery dimers formed by synthetase domains. Based on structural comparison and functional complementation analyses, the active sites of SAICARs and AIRc were identified, including a putative substrate CO(2)-binding site. Furthermore, four symmetry-related, separate tunnel systems in the PAICS octamer were found that connect the active sites of AIRc and SAICARs. This study illustrated the octameric nature of the bifunctional enzyme. Each carboxylase active site is formed by structural elements from three AIRc domains, demonstrating that the octamer structure is essential for the carboxylation activity. Furthermore, the existence of the tunnel system implies a mechanism of intermediate channeling and suggests that the quaternary structure arrangement is crucial for effectively executing the sequential reactions. In addition, this study provides essential structural information for designing PAICS-specific inhibitors for use in cancer chemotherapy.


Asunto(s)
Carboxiliasas/química , Purinas/biosíntesis , Secuencia de Aminoácidos , Sitios de Unión , Carboxiliasas/genética , Carboxiliasas/metabolismo , Cristalografía por Rayos X , Escherichia coli/genética , Prueba de Complementación Genética , Humanos , Luz , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Molecular , Mutagénesis Sitio-Dirigida , Mutación Puntual , Biosíntesis de Proteínas , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Dispersión de Radiación , Homología de Secuencia de Aminoácido , Espectrometría Raman , Difracción de Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA