Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Anal Chem ; 96(28): 11479-11487, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-38943570

RESUMEN

Accurate orientations and stable conformations of membrane receptor immobilization are particularly imperative for accurate drug screening and ligand-protein affinity analysis. However, there remain challenges associated with (1) traditional recombination, purification, and immobilization of membrane receptors, which are time-consuming and labor-intensive; (2) the orientations on the stationary phase are not easily controlled. Herein, a novel one-step synthesis and oriented-immobilization membrane-receptor affinity chromatography (oSOMAC) method was developed to realize high-throughput and accurate drug screening targeting specific domains of membrane receptors. We employed Strep-tag II as a noncovalent immobilization tag fused into platelet-derived growth factor receptor ß (PDGFRß) through CFPS, and meanwhile, the Strep-Tactin-modified monolithic columns are prepared in batches. The advantages of oSOMAC are as follows: (1) targeted membrane receptors can be expressed independent of living cell within 1-2 h; (2) orientation of membrane receptors can be flexibly controlled and active sites can expose accurately; and (3) targeted membrane receptors can be synthesized, purified, and orientation-immobilized on monolithic columns in one step. Accordingly, three potential PDGFRß intracellular domain targeted ligands: tanshinone IIA (Tan IIA), hydroxytanshinone IIA, and dehydrotanshinone IIA were successfully screened out from Salvia miltiorrhiza extract through oSOMAC. Pharmacological experiments and molecular docking further demonstrated that Tan IIA could attenuate hepatic stellate cells activation by targeting the protein kinase domain of PDGFRß with a KD value of 9.7 µM. Ultimately, the novel oSOMAC method provides an original insight for accurate drug screening and interaction analysis which can be applied in other membrane receptors.


Asunto(s)
Receptor beta de Factor de Crecimiento Derivado de Plaquetas , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Ligandos , Humanos , Cromatografía de Afinidad , Evaluación Preclínica de Medicamentos , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/genética , Oligopéptidos/química
2.
Acta Pharmacol Sin ; 44(7): 1429-1441, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36697978

RESUMEN

Aristolochic acid I (AAI) is a well established nephrotoxin and human carcinogen. Cytosolic NAD(P)H quinone oxidoreductase 1 (NQO1) plays an important role in the nitro reduction of aristolochic acids, leading to production of aristoloactam and AA-DNA adduct. Application of a potent NQO1 inhibitor dicoumarol is limited by its life-threatening side effect as an anticoagulant and the subsequent hemorrhagic complications. As traditional medicines containing AAI remain available in the market, novel NQO1 inhibitors are urgently needed to attenuate the toxicity of AAI exposure. In this study, we employed comprehensive 2D NQO1 biochromatography to screen candidate compounds that could bind with NQO1 protein. Four compounds, i.e., skullcapflavone II (SFII), oroxylin A, wogonin and tectochrysin were screened out from Scutellaria baicalensis. Among them, SFII was the most promising NQO1 inhibitor with a binding affinity (KD = 4.198 µmol/L) and inhibitory activity (IC50 = 2.87 µmol/L). In human normal liver cell line (L02) and human renal proximal tubular epithelial cell line (HK-2), SFII significantly alleviated AAI-induced DNA damage and apoptosis. In adult mice, oral administration of SFII dose-dependently ameliorated AAI-induced renal fibrosis and dysfunction. In infant mice, oral administration of SFII suppressed AAI-induced hepatocellular carcinoma initiation. Moreover, administration of SFII did not affect the coagulation function in short term in adult mice. In conclusion, SFII has been identified as a novel NQO1 inhibitor that might impede the risk of AAI to kidney and liver without obvious side effect.


Asunto(s)
Ácidos Aristolóquicos , Ratones , Humanos , Animales , Ácidos Aristolóquicos/toxicidad , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Riñón/patología , Hígado/metabolismo
3.
Drug Chem Toxicol ; 46(2): 304-313, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35057680

RESUMEN

Salvianolic acid A (SA-A), a water-soluble compound extracted from traditional Chinese herb Radix Salvia miltiorrhiza, has anti-fibrotic effects on carbon tetrachloride (CCl4)-induced liver fibrosis. However, the underlying molecular mechanism remains unclear. Thus, this study aimed to elucidate the molecular mechanism underlying the anti-fibrotic effects of SA-A on CCl4-induced liver fibrosis in mice. All mice (except control group) were intraperitoneally administered CCl4 dissolved in peanut oil to induce liver fibrosis. Treatment groups were then gavaged with SA-A (20 or 40 mg/kg). The liver function index; liver fibrosis index; and superoxide dismutase (SOD), malondialdehyde (MDA), and glutathione peroxidase (GSH-Px) levels were determined. Furthermore, histopathological changes in liver tissues were observed via hematoxylin-eosin and Masson's trichrome staining. The expression of α-smooth muscle actin (α-SMA) and collagen I was detected using immunofluorescence, and the mRNA levels of inflammatory factors were determined using quantitative polymerase chain reaction. Finally, western blotting and immunofluorescence were used to determine the expression levels of proteins related to Nrf2/HO-1, NF-κB/IκBα, p38 MAPK, and JAK1/STAT3 signaling pathways. The results showed that SA-A could ameliorate CCl4-induced liver injury and liver fibrosis, improve morphology, and alleviate collagen deposition in the fibrotic liver. Moreover, SA-A could regulate the Nrf2/HO-1, NF-κB/IκBα, p38 MAPK, and JAK1/STAT3 signaling pathways; increase the levels of SOD and GSH-Px; and decrease MDA level in the fibrotic liver. Collectively, our study findings indicate that SA-A is effective in preventing liver fibrosis in mice by inhibiting inflammation and oxidative stress via regulating the Nrf2/HO-1, NF-κB/IκBα, p38 MAPK, and JAK1/STAT3 signaling pathways.


Asunto(s)
Factor 2 Relacionado con NF-E2 , FN-kappa B , Ratas , Ratones , Animales , FN-kappa B/metabolismo , FN-kappa B/farmacología , Inhibidor NF-kappaB alfa/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Ratas Sprague-Dawley , Transducción de Señal , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/prevención & control , Hígado , Estrés Oxidativo , Superóxido Dismutasa/metabolismo , Tetracloruro de Carbono/toxicidad
4.
J Cell Mol Med ; 24(9): 5122-5134, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32185887

RESUMEN

Angelica sinensis (AS; Dang Gui), a traditional Chinese herb, has for centuries been used for the treatment of bone diseases, including osteoporosis and osteonecrosis. However, the effective ingredient and underlying mechanisms remain elusive. Here, we identified guaiacol as the active component of AS by two-dimensional cell membrane chromatography/C18 column/time-of-flight mass spectrometry (2D CMC/C18 column/TOFMS). Guaiacol suppressed osteoclastogenesis and osteoclast function in bone marrow monocytes (BMMCs) and RAW264.7 cells in vitro in a dose-dependent manner. Co-immunoprecipitation indicated that guaiacol blocked RANK-TRAF6 association and RANK-C-Src association. Moreover, guaiacol prevented phosphorylation of p65, p50, IκB (NF-κB pathway), ERK, JNK, c-fos, p38 (MAPK pathway) and Akt (AKT pathway), and reduced the expression levels of Cathepsin K, CTR, MMP-9 and TRAP. Guaiacol also suppressed the expression of nuclear factor of activated T-cells cytoplasmic 1(NFATc1) and the RANKL-induced Ca2+ oscillation. In vivo, it ameliorated ovariectomy-induced bone loss by suppressing excessive osteoclastogenesis. Taken together, our findings suggest that guaiacol inhibits RANKL-induced osteoclastogenesis by blocking the interactions of RANK with TRAF6 and C-Src, and by suppressing the NF-κB, MAPK and AKT signalling pathways. Therefore, this compound shows therapeutic potential for osteoclastogenesis-related bone diseases, including postmenopausal osteoporosis.


Asunto(s)
Proteína Tirosina Quinasa CSK/metabolismo , Guayacol/farmacología , Osteoclastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Adipogénesis , Animales , Células de la Médula Ósea/citología , Resorción Ósea , Proliferación Celular , Femenino , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Subunidad p50 de NF-kappa B/antagonistas & inhibidores , Osteoclastos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células RAW 264.7 , Microtomografía por Rayos X
5.
J Cell Mol Med ; 24(16): 9067-9084, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32604472

RESUMEN

Psoralea corylifolia (P corylifolia) has been popularly applied in traditional Chinese medicine decoction for treating osteoporosis and promoting fracture healing since centuries ago. However, the bioactive natural components remain unknown. In this study, applying comprehensive two-dimensional cell membrane chromatographic/C18 column/time-of-flight mass spectrometry (2D CMC/C18 column/TOFMS) system, neobavaisoflavone (NBIF), for the first time, was identified for the bioaffinity with RAW 264.7 cells membranes from the extracts of P corylifolia. Here, we revealed that NBIF inhibited RANKL-mediated osteoclastogenesis in bone marrow monocytes (BMMCs) and RAW264.7 cells dose dependently at the early stage. Moreover, NBIF inhibited osteoclasts function demonstrated by actin ring formation assay and pit-formation assay. With regard to the underlying molecular mechanism, co-immunoprecipitation showed that both the interactions of RANK with TRAF6 and with c-Src were disrupted. In addition, NBIF inhibited the phosphorylation of P50, P65, IκB in NF-κB pathway, ERK, JNK, P38 in MAPKs pathway, AKT in Akt pathway, accompanied with a blockade of calcium oscillation and inactivation of nuclear translocation of nuclear factor of activated T cells cytoplasmic 1 (NFATc1). In vivo, NBIF inhibited osteoclastogenesis, promoted osteogenesis and ameliorated bone loss in ovariectomized mice. In summary, P corylifolia-derived NBIF inhibited RANKL-mediated osteoclastogenesis by suppressing the recruitment of TRAF6 and c-Src to RANK, inactivating NF-κB, MAPKs, and Akt signalling pathways and inhibiting calcium oscillation and NFATc1 translocation. NBIF might serve as a promising candidate for the treatment of osteoclast-associated osteopenic diseases.


Asunto(s)
Genes src/efectos de los fármacos , Isoflavonas/farmacología , Osteogénesis/efectos de los fármacos , Ligando RANK/metabolismo , Transducción de Señal/efectos de los fármacos , Factor 6 Asociado a Receptor de TNF/metabolismo , Animales , Médula Ósea/efectos de los fármacos , Médula Ósea/metabolismo , Resorción Ósea/tratamiento farmacológico , Resorción Ósea/metabolismo , Línea Celular , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Monocitos/efectos de los fármacos , Monocitos/metabolismo , FN-kappa B/metabolismo , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células RAW 264.7
6.
Xenobiotica ; 49(8): 975-980, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30215539

RESUMEN

1. In clinical practice, diammonium glycyrrhizinate is usually used with omeprazole in patients with viral hepatitis and cirrhosis accompanied by peptic ulcers. However, the drug interaction between diammonium glycyrrhizinate and omeprazole remains unclear. 2. In this study, the effects of diammonium glycyrrhizinate on the pharmacokinetics of omeprazole was investigated by liquid chromatography-tandem mass spectrometry (LC-MS/MS) analytical method. Male Sprague-Dawley rats were randomly assigned to two groups: omeprazole and omeprazole + diammonium glycyrrhizinate, and the main pharmacokinetic parameters were estimated after oral administration. It was found that using the omeprazole along with the diammonium glycyrrhizinate increased the AUC, AUMC, Cmax for omeprazole. 3. For this reason, we used the LC-MS/MS to detect the binding rate of plasma protein (BRPP) of omeprazole in rats, it was found that diammonium glycyrrhizinate could decrease the BRPP in rats. In addition, we found that diammonium glycyrrhizinate specifically inhibited the enzyme activity of the CYP2C19 and CYP3A4, which are involved in the metabolism of the omeprazole. 4. These results mean that diammonium glycyrrhizinate could inhibit the metabolism and increase the plasma concentration of the omeprazole in rats. Overall, diammonium glycyrrhizinate can influence the pharmacokinetics of omeprazole by inhibiting CYP2C19 and CYP3A4 activities and decreasing BRPP of omeprazole.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Ácido Glicirrínico/farmacología , Omeprazol/farmacocinética , Animales , Estudios de Factibilidad , Masculino , Unión Proteica/efectos de los fármacos , Ratas Sprague-Dawley , Estándares de Referencia
7.
Chin J Nat Med ; 21(7): 483-498, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37517817

RESUMEN

Insomnia is a common sleep disorder without effective therapy and can affect a person's life. The mechanism of the disease is not completely understood. Hence, there is a need to understand the targets related to insomnia, in order to develop innovative therapies and new compounds. Recently, increasing interest has been focused on complementary and alternative medicines for treating or preventing insomnia. Research into their molecular components has revealed that their sedative and sleep-promoting properties rely on the interactions with various neurotransmitter systems in the brain. In this review, the role of 5-hydroxytryptamine (5-HT) in insomnia development is summarized, while a systematic analysis of studies is conducted to assess the mechanisms of herbal medicines on different 5-HT receptors subtypes, in order to provide reference for subsequent research.


Asunto(s)
Plantas Medicinales , Trastornos del Inicio y del Mantenimiento del Sueño , Trastornos del Inicio y del Mantenimiento del Sueño/tratamiento farmacológico , Trastornos del Inicio y del Mantenimiento del Sueño/metabolismo , Receptores de Serotonina/metabolismo , Serotonina/metabolismo , Sueño , Humanos , Animales
8.
Int J Biol Macromol ; 235: 123639, 2023 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-36822287

RESUMEN

Platelet-derived growth factor receptor ß (PDGFRß) plays an important role in hepatic fibrosis and is closely associated with hepatic stellate cells (HSCs) activation. Previously, by modeling PDGFRß affinity chromatography, we found that gomisin D can target PDGFRß. However, whether gomisin D has anti-fibrosis effects through targeting PDGFRß remained unclear. In this study, the effect of gomisin D on hepatic fibrosis was evaluated in vivo and vitro. HSC cell lines and primary HSC were cultured and functionally we found that gomisin D promotes HSC apoptosis, inhibits HSCs activation and proliferation. A male BALB/c mouse liver fibrosis model was established to comfirm gomisin D (especially in 50 mg/kg) could improve liver fibrosis by inhibiting HSCs activation. In addition, gomisin D had a good binding ability with PDGFRß (KD = 3.3e-5 M). Mechanically, gomisin D regulated PDGF-BB/PDGFRß signaling pathway by targeting PDGFRß, further more inhibited HSC activation, subsequently inhibited inflammatory factors, ultimately improved CCl4-induced liver fibrosis. Overall, gomisin D could inhibit HSC proliferation and activation, promote HSC apoptosis, and alleviate CCl4-induced hepatic fibrosis by targeting PDGFRß and regulating PDGF-BB/PDGFRß signaling pathway. This study provides a new drug for anti-liver firbosis therapy, and elucidates the deeper mechanism of gomisin D against HSCs activation by targeting PDGFRß.


Asunto(s)
Indoles , Lignanos , Hígado , Schisandra , Schisandra/química , Lignanos/farmacología , Indoles/farmacología , Masculino , Ratones , Animales , Ratones Endogámicos BALB C , Células Madre Hematopoyéticas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proliferación Celular/efectos de los fármacos , Apoptosis/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Fibrosis/metabolismo , Tetracloruro de Carbono , Transducción de Señal , Humanos
9.
Front Pharmacol ; 14: 1092151, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37288106

RESUMEN

Liver fibrosis is considered a sustained wound healing response and metabolic syndrome, and its therapy is of great significance for chronic liver disease. Schizandrin C, as one lignan from hepatic protectant Schisandra chinensis, can depress the oxidative effect and lipid peroxidation, and protect against liver injury. In this study, C57BL/6J mice were used to estimate a liver fibrosis model by CCl4, and Schizandrin C exerted an anti-hepatic fibrosis effect, as evidenced by decreased alanine aminotransferase, aspartate aminotransferase and total bilirubin activities in serum, lower hydroxyproline content, recuperative structure and less collagen accumulation in the liver. In addition, Schizandrin C reduced the expressions of alpha-smooth muscle actin and type Ι collagen in the liver. In vitro experiments also revealed that Schizandrin C attenuated hepatic stellate cell activation in both LX-2 and HSC-T6 cells. Furthermore, lipidomics and quantitative real-time PCR analysis revealed that Schizandrin C regulated the lipid profile and related metabolic enzymes in the liver. In addition, the mRNA levels of inflammation factors were downregulated by Schizandrin C treatment, accompanied by lower protein levels of IκB-Kinase-ß, nuclear factor kappa-B p65, and phospho-nuclear factor kappa-B p65. Finally, Schizandrin C inhibited the phosphorylation of p38 MAP kinase and extracellular signal-regulated protein kinase, which were activated in the CCl4 fibrotic liver. Taken together, Schizandrin C can regulate lipid metabolism and inflammation to ameliorate liver fibrosis by nuclear factor kappa-B and p38/ERK MAPK signaling pathways. These findings supported Schizandrin C as a potential drug for liver fibrosis.

10.
Int Immunopharmacol ; 122: 110550, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37451016

RESUMEN

Liver fibrosis is a reversible pathological process and a wound healing response to liver injury. As an early stage of various liver diseases, liver fibrosis can develop into cirrhosis, liver failure, and even liver cancer if not controlled in time. Salvia miltiorrhiza is a medicinal plant with hepatoprotective effects. Salvianolic acid B (Sal B) is the representative component of S. miltiorrhiza. Many studies have reported the anti-liver fibrosis effects and mechanisms of Sal B. However, the direct anti-fibrotic targets of Sal B have not yet been reported. Platelet-derived growth factor receptor ß (PDGFRß) is one of the most classical targets in liver fibrosis, which is closely related to hepatic stellate cells (HSCs) activated. Previously, we established and applied a PDGFRß affinity chromatography model, and found that Sal B binds well to PDGFRß. Therefore, this study aimed to investigate the direct targets of Sal B against liver fibrosis. We confirmed the binding ability of Sal B to PDGFRß by molecular docking and a surface plasmon resonance biosensor. Our findings indicated that Sal B targeted PDGFRß to inhibit the activation, migration and proliferation of HSCs and suppressed the PDGF-BB-induced PDGFRß signaling pathway. Annexin V-FITC/PI assay showed that Sal B reversed the PDGF-BB-induced decrease in HSC apoptosis rate. In the mouse liver fibrosis model, Sal B inhibited the PDGFRß signaling pathway, HSC activation and reduced inflammatory response, ultimately improved CCl4-induced liver fibrosis. In summary, the direct anti-fibrotic targets of Sal B may be PDGFRß, and this study clarified the anti-liver fibrosis effects and mechanism of Sal B.


Asunto(s)
Células Estrelladas Hepáticas , Cirrosis Hepática , Ratones , Animales , Becaplermina/farmacología , Simulación del Acoplamiento Molecular , Cirrosis Hepática/metabolismo , Fibrosis
11.
Phytomedicine ; 107: 154435, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36155216

RESUMEN

BACKGROUND: Long non-coding RNA (LncRNAs) have been reported to play an important role in liver fibrosis and are closely associated with hepatic stellate cell (HSC) activation. We previously found that salvianolic acid B (Sal B) improves liver fibrosis by regulating the NF-κB signaling pathway. However, whether the LncRNA, regulator of reprogramming (LncRNA-ROR) plays a role in Sal B-mediated anti-fibrosis effects via the NF-κB signaling pathway remain unclear. PURPOSE: This study aimed to evaluate the effects of Sal B on HSC activation and liver fibrosis and investigate its mechanism from the perspective of LncRNA-ROR-mediated NF-κB signaling pathways. METHODS: LX-2 and T6 cell lines were cultured. Animal models of liver fibrosis were established using CCl4 in male BALB/c mice. Primary HSCs were isolated from mice and cultured. Serum biochemical and liver histological analyses were performed to evaluate the effects of Sal B on liver fibrosis. The index of HSC activation and the expression of LncRNA-ROR, microRNAs (miRNAs), and inflammatory factors were determined by quantitative real-time reverse transcription-polymerase chain reaction (qRT-PCR) or immunofluorescence staining. Cell proliferation was measured by a Cell Counting Kit-8 (CCK-8). NF-κB signaling-associated protein levels were assessed using western blotting or immunofluorescence staining. A luciferase reporter assay was used to detect transcription activity. RESULTS: In this study, a lower level of LncRNA-ROR was found during Sal B attenuating HSC activation in HSCs. Mechanistically, Sal B impeded the NF-κB signaling pathway to inhibit HSC proliferation and activation by downregulating LncRNA-ROR. Additionally, Sal B upregulated miR-6499-3p to target LncRNA-ROR for degradation. Functionally, Sal B treatment ameliorated CCl4-induced liver fibrosis in mice by inhibiting HSC activation. CONCLUSION: Sal B suppresses HSC activation and liver fibrosis via regulation of miR-6499-3p/LncRNA-ROR-mediated NF-κB signaling pathway. These results reveal a new molecular mechanism of Sal B on liver fibrosis from the insight of LncRNAs.


Asunto(s)
MicroARNs , ARN Largo no Codificante , Animales , Benzofuranos , Depsidos , Células Estrelladas Hepáticas , Cirrosis Hepática/metabolismo , Masculino , Ratones , MicroARNs/genética , MicroARNs/metabolismo , FN-kappa B/metabolismo , ARN Largo no Codificante/genética , Transducción de Señal
12.
J Pharm Anal ; 12(5): 725-732, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36320599

RESUMEN

Astragali Radix (AR) is a clinically used herbal medicine with multiple immunomodulatory activities that can strengthen the activity and cytotoxicity of natural killer (NK) cells. However, owing to the complexity of its composition, the specific active ingredients in AR that act on NK cells are not clear yet. Cell membrane chromatography (CMC) is mainly used to screen the active ingredients in a complex system of herbal medicines. In this study, a new comprehensive two-dimensional (2D) NK-92MI CMC/C18 column/time-of-flight mass spectrometry (TOFMS) system was established to screen for potential NK cell activators. To obtain a higher column efficiency, 3-mercaptopropyltrimethoxysilane-modified silica was synthesized to prepare the NK-92MI CMC column. In total, nine components in AR were screened from this system, which could be washed out from the NK-92MI/CMC column after 10 min, and they showed good affinity for NK-92MI/CMC column. Two representative active compounds of AR, isoastragaloside I and astragaloside IV, promoted the killing effect of NK cells on K562 cells in a dose-dependent manner. It can thus suggest that isoastragaloside I and astragaloside IV are the main immunomodulatory components of AR. This comprehensive 2D NK-92MI CMC analytical system is a practical method for screening immune cell activators from other herbal medicines with immunomodulatory effects.

13.
Acta Pharm Sin B ; 12(9): 3682-3693, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36176904

RESUMEN

Cell membrane affinity chromatography has been widely applied in membrane protein (MP)-targeted drug screening and interaction analysis. However, in current methods, the MP sources are derived from cell lines or recombinant protein expression, which are time-consuming for cell culture or purification, and also difficult to ensure the purity and consistent orientation of MPs in the chromatographic stationary phase. In this study, a novel in situ synthesis membrane protein affinity chromatography (iSMAC) method was developed utilizing cell-free protein expression (CFE) and covalent immobilized affinity chromatography, which achieved efficient in situ synthesis and unidirectional insertion of MPs into liposomes in the stationary phase. The advantages of iSMAC are: 1) There is no need to culture cells or prepare recombinant proteins; 2) Specific and purified MPs with stable and controllable content can be obtained within 2 h; 3) MPs maintain the transmembrane structure and a consistent orientation in the chromatographic stationary phase; 4) The flexible and personalized construction of cDNAs makes it possible to analyze drug binding sites. iSMAC was successfully applied to screen PDGFRß inhibitors from Salvia miltiorrhiza and Schisandra chinensis. Micro columns prepared by in-situ synthesis maintain satisfactory analysis activity within 72 h. Two new PDGFRß inhibitors, salvianolic acid B and gomisin D, were screened out with K D values of 13.44 and 7.39 µmol/L, respectively. In vitro experiments confirmed that the two compounds decreased α-SMA and collagen Ӏ mRNA levels raised by TGF-ß in HSC-T6 cells through regulating the phosphorylation of p38, AKT and ERK. In vivo, Sal B could also attenuate CCl4-induced liver fibrosis by downregulating PDGFRß downstream related protein levels. The iSMAC method can be applied to other general MPs, and provides a practical approach for the rapid preparation of MP-immobilized or other biological solid-phase materials.

14.
Clin Transl Med ; 12(11): e1095, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36314067

RESUMEN

BACKGROUND: Transketolase (TKT), a key rate-limiting enzyme in the non-oxidative branch of the pentose phosphate pathway (PPP), provides more than 85% of the ribose required for de novo nucleotide biosynthesis and promotes the development of hepatocellular carcinoma (HCC). Pharmacologic inhibition of TKT could impede HCC development and enhance treatment efficacy. However, no safe and effective TKT inhibitor has been approved. METHODS: An online two-dimensional TKT protein immobilised biochromatographic system was established for high-throughput screening of TKT ligands. Oroxylin A was found to specifically bind TKT. Drug affinity responsive target stability, cellular thermal shift assay, surface plasmon resonance, molecular docking, competitive displacement assay, and site mutation were performed to identify the binding of oroxylin A with TKT. Antitumour effects of oroxylin A were evaluated in vitro, in human xenograft mice, diethylnitrosamine (DEN)-induced HCC mice, and patient-derived organoids (PDOs). Metabolomic analysis was applied to detect the enzyme activity. Transcriptome profiling was conducted to illustrate the anti-HCC mechanism of oroxylin A. TKT knocking-down HCC cell lines and PDOs were established to evaluate the role of TKT in oroxylin A-induced HCC suppression. RESULTS: By targeting TKT, oroxylin A stabilised the protein to proteases and temperature extremes, decreased its activity and expression, resulted in accumulation of non-oxidative PPP substrates, and activated p53 signalling. In addition, oroxylin A suppressed cell proliferation, induced apoptosis and cell-cycle arrest, and inhibited the growth of human xenograft tumours and DEN-induced HCC in mice. Crucially, TKT depletion exerted identical effects to oroxylin A, and the promising inhibitor also exhibited excellent therapeutic efficacy against clinically relevant HCC PDOs. CONCLUSIONS: These results uncover a unique role for oroxylin A in TKT inhibition, which directly targets TKT and suppresses the non-oxidative PPP. Our findings will facilitate the development of small-molecule inhibitors of TKT and novel therapeutics for HCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Animales , Ratones , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Transcetolasa/genética , Transcetolasa/metabolismo , Vía de Pentosa Fosfato , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Organoides/metabolismo , Organoides/patología , Simulación del Acoplamiento Molecular
15.
Artículo en Inglés | MEDLINE | ID: mdl-33418242

RESUMEN

Screening active components targeting membrane proteins is important for drug discovery from traditional Chinese medicine. Cell membrane chromatography (CMC) has achieved a wide application in screening active components on pathological cells due to its high sensitivity and effectiveness. However, it is hard to clarify the specific target protein through simply using pathological and normal cells. In this study, a novel comparative two-dimensional (2D) cell membrane chromatography system was established. Based on the construction of hepatocellular carcinoma cell line SK-Hep1-GPC3 with high expression of protein Glypican-3 (GPC3), SK-Hep1-GPC3/CMC column was loaded to screen selective antitumor components from Scutellariae Radix according to the retention behaviors on column. Viscidulin I was retained on SK-Hep1-GPC3/CMC column, and showed 4.33 µM affinity to GPC3 according to surface plasmon resonance (SPR). The IC50 of viscidulin I on SK-Hep1-GPC3 cells was 18.01 µM in cell proliferation assay. Thus, this method can be applied to screen complex herbal medicines for ligands bound to specific target protein receptor related to hepatic carcinoma.


Asunto(s)
Antineoplásicos , Membrana Celular/metabolismo , Cromatografía de Afinidad/métodos , Glipicanos , Scutellaria baicalensis/química , Antineoplásicos/análisis , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Línea Celular Tumoral , Descubrimiento de Drogas , Medicamentos Herbarios Chinos/análisis , Medicamentos Herbarios Chinos/metabolismo , Medicamentos Herbarios Chinos/farmacología , Flavonoides/análisis , Flavonoides/metabolismo , Flavonoides/farmacología , Glipicanos/antagonistas & inhibidores , Glipicanos/metabolismo , Humanos , Neoplasias Hepáticas/metabolismo , Reproducibilidad de los Resultados
16.
J Pharm Biomed Anal ; 195: 113825, 2021 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-33339641

RESUMEN

Crystal-induced kidney injury (CIKI) is the fundamental pathological change during nephrolithiasis, although the molecular mechanism is still unclear. Pyrrosia calvata (Bak.) Ching has been used in folk medicine to treat urolithiasis for years. To clarify the pharmacodynamic substances and the mechanism of its antiurolithiasis effects, in this study, a novel, stop-flow, comprehensive, two-dimensional (2D) HK-2 and HK-2/CIKI cell membrane chromatography (CMC) comparative analysis system was developed to screen for the potential active ingredients from Pyrrosia calvata (Bak.) Ching against CIKI. The comprehensive 2D CMC comparative analysis system showed satisfactory selectivity, and eight ingredients were screened and identified by this system. Among them, mangiferin exhibited higher affinity for the HK-2/CIKI CMC column than the HK-2 CMC column and was selected for further efficacy verification. Cell proliferation assays showed that mangiferin could protect HK-2 cell viability after stimulation with sodium oxalate (NaOX). Additionally, in a rodent model of CIKI, mangiferin decreased the deposition of calcium oxalate (CaOX) crystals in mouse kidneys, alleviated the pathological damage to kidney tissue, and inhibited the upregulation of OPN, MCP1, and CD44 expression caused by CaOX crystals. The established comprehensive 2D CMC comparative analysis system can be applied to screen active ingredients with disease specificity from traditional Chinese medicine (TCM) and is suitable for other cell models.


Asunto(s)
Oxalato de Calcio , Riñón , Animales , Membrana Celular , Supervivencia Celular , Cromatografía , Ratones
17.
Acta Pharm Sin B ; 11(1): 222-236, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33072499

RESUMEN

Lianhuaqingwen (LHQW) capsule, a herb medicine product, has been clinically proved to be effective in coronavirus disease 2019 (COVID-19) pneumonia treatment. However, human exposure to LHQW components and their pharmacological effects remain largely unknown. Hence, this study aimed to determine human exposure to LHQW components and their anti-COVID-19 pharmacological activities. Analysis of LHQW component profiles in human plasma and urine after repeated therapeutic dosing was conducted using a combination of HRMS and an untargeted data-mining approach, leading to detection of 132 LHQW prototype and metabolite components, which were absorbed via the gastrointestinal tract and formed via biotransformation in human, respectively. Together with data from screening by comprehensive 2D angiotensin-converting enzyme 2 (ACE2) biochromatography, 8 components in LHQW that were exposed to human and had potential ACE2 targeting ability were identified for further pharmacodynamic evaluation. Results show that rhein, forsythoside A, forsythoside I, neochlorogenic acid and its isomers exhibited high inhibitory effect on ACE2. For the first time, this study provides chemical and biochemical evidence for exploring molecular mechanisms of therapeutic effects of LHQW capsule for the treatment of COVID-19 patients based on the components exposed to human. It also demonstrates the utility of the human exposure-based approach to identify pharmaceutically active components in Chinese herb medicines.

18.
ACS Appl Bio Mater ; 3(8): 5000-5006, 2020 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-35021677

RESUMEN

Cell membrane chromatography (CMC) has been widely used for characterizing the interaction between drugs and membrane receptors to screen target components from herbal medicines. However, the column life, stability, and the efficiency cannot meet the needs of high-throughput screening purpose. In this study, a P-glycoprotein immobilized cell membrane stationary phase (P-gp/CMSP) was prepared with a simple and mild two-step aldehyde modification, realizing the covalent bonding between cell membrane and stationary phase. The column life and stability were significantly enhanced compared with the unmodified columns. The P-gp/CMC column was equipped into a comprehensive 2D P-gp/CMC/Capcell-C18/TOFMS system, which actualizes the automated and high-throughput analytical process and rapid identification of complex chemical samples with no data loss. Five compounds with significant retention were screened out and unambiguously identified by the comprehensive 2D analytical system. Baicalin was confirmed as a P-gp inhibitor with ATP depletion inhibition ratio of 83.4%. Moreover, the reversal index of baicalin on DOX significantly increased to 11.13 when its concentration reached 25 µM, revealing that baicalin could effectively reverse the MDR cell model induced by DOX. The integrated system is a practical drug discovery platform and could be applied to other transmembrane protein models.

19.
Acta Pharm Sin B ; 10(10): 1856-1865, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33163340

RESUMEN

Osteoporosis is a bone metabolic disease caused by the imbalance between osteoblasts and osteoclasts due to excess osteoclastogenesis, manifesting in the decrease of bone density and bone strength. Scutellariae Radix shows good anti-osteoporosis activity, but the effective component is still unclear. Cell membrane chromatography (CMC) is a biological affinity chromatography with membrane immobilized on a silica carrier as the stationary phase. It can realize a dynamical simulation of interactions between drugs and receptors on cell membrane, which is suitable for screening active compounds from complex systems. In this study, the components of Scutellariae Radix with potential anti-osteoporosis activity through inhibiting the differentiation from bone marrow mononuclear cells (BMMCs) to osteoclast were screened by a BMMC/CMC analytical system. Firstly, a new 3-mercaptopropyltrimethoxysilane (MPTS)-modified BMMC/CMC stationary phase was developed to realize covalent binding with cell membrane fractions. By investigating the retention time (t R) of the positive drug, the life span of the MPTS-modified CMC columns was significantly improved from 3 to 12 days. Secondly, 6 components of Scutellariae Radix were screened to show affinity to membrane receptors on BMMCs by a two-dimensional BMMC/CMC-TOFMS analytical system. Among them, tectochrysin demonstrated the best anti-osteoporosis effect in vitro, which has never been reported. We found that tectochrysin could inhibit the differentiation of BMMCs into osteoclasts induced by receptor activator of nuclear factor-κΒ ligand (RANKL) and macrophage colony-stimulating factor (M-CSF) in a concentration-dependent manner in vitro. In vivo, it significantly reduced the loss of bone trabeculae in ovariectomized mice, and decreased the level of C-terminal cross-linking telopeptides of type 1 collagen (CTX-1), tartrate-resistant acid phosphatase 5b (TRAP-5b), interleukin 6 (IL-6) in serum. In conclusion, tectochrysin serves as a potential candidate in the treatment of osteoporosis. The proposed two-dimensional MPTS-modified BMMC/CMC-TOFMS analytical system shows the advantages of long-life span and fast recognition ability, which is very suitable for infrequent cell lines.

20.
Sci Adv ; 6(47)2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33208358

RESUMEN

The actin-bundling protein L-plastin (LPL) mediates the resorption activity of osteoclasts, but its therapeutic potential in pathological bone loss remains unexplored. Here, we report that LPL knockout mice show increased bone mass and cortical thickness with more mononuclear tartrate-resistant acid phosphatase-positive cells, osteoblasts, CD31hiEmcnhi endothelial vessels, and fewer multinuclear osteoclasts in the bone marrow and periosteum. LPL deletion impeded preosteoclasts fusion by inhibiting filopodia formation and increased the number of preosteoclasts, which release platelet-derived growth factor-BB to promote CD31hiEmcnhi vessel growth and bone formation. LPL expression is regulated by the phosphatidylinositol 3-kinase/AKT/specific protein 1 axis in response to receptor activator of nuclear factor-κB ligand. Furthermore, we identified an LPL inhibitor, oroxylin A, that could maintain bone mass in ovariectomy-induced osteoporosis and accelerate bone fracture healing in mice. In conclusion, we showed that LPL regulates osteoclasts fusion, and targeting LPL serves as a novel anabolic therapy for pathological bone loss.


Asunto(s)
Actinas , Osteoclastos , Actinas/metabolismo , Animales , Femenino , Glicoproteínas de Membrana , Ratones , Proteínas de Microfilamentos , Osteoclastos/metabolismo , Osteogénesis , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA