Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Ethnopharmacol ; 328: 118116, 2024 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-38548118

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Aristolochic acids (AAs) are naturally occurring nitro phenanthrene carboxylic acids primarily found in plants of the Aristolochiaceae family. Aristolochic acid D (AAD) is a major constituent in the roots and rhizomes of the Chinese herb Xixin (the roots and rhizomes of Asarum heterotropoides F. Schmidt), which is a key material for preparing a suite of marketed Chinese medicines. Structurally, AAD is nearly identical to the nephrotoxic aristolochic acid I (AAI), with an additional phenolic group at the C-6 site. Although the nephrotoxicity and metabolic pathways of AAI have been well-investigated, the metabolic pathway(s) of AAD in humans and the influence of AAD metabolism on its nephrotoxicity has not been investigated yet. AIM OF THE STUDY: To identify the major metabolites of AAD in human tissues and to characterize AAD O-glucuronidation kinetics in different enzyme sources, as well as to explore the influence of AAD O-glucuronidation on its nephrotoxicity. MATERIALS AND METHODS: The O-glucuronide of AAD was biosynthesized and its chemical structure was fully characterized by both 1H-NMR and 13C-NMR. Reaction phenotyping assays, chemical inhibition assays, and enzyme kinetics analyses were conducted to assess the crucial enzymes involved in AAD O-glucuronidation in humans. Docking simulations were performed to mimic the catalytic conformations of AAD in human UDP-glucuronosyltransferases (UGTs), while the predicted binding energies and distances between the deprotonated C-6 phenolic group of AAD and the glucuronyl moiety of UDPGA in each tested human UGT isoenzyme were measured. The mitochondrial membrane potentials (MMP) and reactive oxygen species (ROS) levels in HK-2 cells treated with either AAI, or AAD, or AAD O-glucuronide were tested, to elucidate the impact of O-glucuronidation on the nephrotoxicity of AAD. RESULTS: AAD could be rapidly metabolized in human liver and intestinal microsomes (HLM and HIM, respectively) to form a mono-glucuronide, which was purified and fully characterized as AAD-6-O-ß-D-glucuronide (AADG) by NMR. UGT1A1 was the predominant enzyme responsible for AAD-6-O-glucuronidation, while UGT1A9 contributed to a lesser extent. AAD-6-O-glucuronidation in HLM, HIM, UGT1A1 and UGT1A9 followed Michaelis-Menten kinetics, with the Km values of 4.27 µM, 9.05 µM, 3.87 µM, and 7.00 µM, respectively. Docking simulations suggested that AAD was accessible to the catalytic cavity of UGT1A1 or UGT1A9 and formed catalytic conformations. Further investigations showed that both AAI and AAD could trigger the elevated intracellular ROS levels and induce mitochondrial dysfunction and in HK-2 cells, but AADG was hardly to trigger ROS accumulation and mitochondrial dysfunction. CONCLUSION: Collectively, UGT1A-catalyzed AAD 6-O-glucuronidation represents a crucial detoxification pathway of this naturally occurring AAI analogs in humans, which is very different from that of AAI.


Assuntos
Ácidos Aristolóquicos , Doenças Mitocondriais , Humanos , Ácidos Aristolóquicos/toxicidade , Glucuronídeos/metabolismo , Microssomos Hepáticos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Glucuronosiltransferase/metabolismo , Cinética , Catálise , Difosfato de Uridina/metabolismo
2.
Phytomedicine ; 126: 155458, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38394733

RESUMO

BACKGROUND: As a canonical iron-dependent form of regulated cell death (RCD), ferroptosis plays a crucial role in chemical-induced liver injuries. Previous studies have demonstrated that xanthohumol (Xh), a natural prenylflavonoid isolated from hops, exhibits anti-inflammatory, anti-antioxidative and hepatoprotective properties. However, the regulatory effects of Xh on hepatic ferroptosis and the underlying mechanism have not yet been fully elucidated. PURPOSE: To investigate the hepatoprotective effects of Xh against drug-induced liver injury (DILI) and the regulatory effects of Xh on hepatic ferroptosis, as well as to reveal the underlying molecular mechanisms. METHODS/STUDY DESIGN: The hepatoprotective benefits of Xh were investigated in APAP-induced liver injury (AILI) mice and HepaRG cells. Xh was administered intraperitoneally to assess its in vivo effects. Histological and biochemical studies were carried out to evaluate liver damage. A series of ferroptosis-related markers, including intracellular Fe2+ levels, ROS and GSH levels, the levels of MDA, LPO and 4-HNE, as well as the expression levels of ferroptosis-related proteins and modulators were quantified both in vivo and in vitro. The modified peptides of Keap1 by Xh were characterized utilizing nano LC-MS/MS. RESULTS: Xh remarkably suppresses hepatic ferroptosis and ameliorates AILI both in vitro and in vivo, via suppressing Fe2+ accumulation, ROS formation, MDA generation and GSH depletion, these observations could be considerably mitigated by the ferroptosis inhibitor ferrostatin-1 (Fer-1). Mechanistically, Xh could significantly activate the Nrf2/xCT/GPX4 signaling pathway to counteract AILI-induced hepatocyte ferroptosis. Further investigations showed that Xh could covalently modify three functional cysteine residues (cys151, 273, 288) of Keap1, which in turn, reduced the ubiquitination rates of Nrf2 and prolonged its degradation half-life. CONCLUSIONS: Xh evidently suppresses hepatic ferroptosis and ameliorates AILI via covalent modifying three key cysteines of Keap1 and activating Nrf2/xCT/GPX4 signaling pathway.


Assuntos
Ferroptose , Flavonoides , Propiofenonas , Animais , Camundongos , Proteína 1 Associada a ECH Semelhante a Kelch , Fator 2 Relacionado a NF-E2 , Espécies Reativas de Oxigênio , Espectrometria de Massas em Tandem , Fígado , Transdução de Sinais , Cisteína
4.
Anal Chem ; 95(42): 15665-15672, 2023 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-37782032

RESUMO

Human carboxylesterase 2A (hCES2A) is an important endoplasmic reticulum (ER)-resident enzyme that is responsible for the hydrolytic metabolism or activation of numerous ester-bearing drugs and environmental toxins. The previously reported hCES2A fluorogenic substrates suffer from limited emission wavelength, low specificity, and poor localization accuracy, thereby greatly limiting the in situ functional imaging of hCES2A and drug discovery. Herein, a rational ligand design strategy was adopted to construct a highly specific near-infrared (NIR) substrate for hCES2A. Following scaffold screening and recognition group optimization, HTCF was identified as a desirable NIR fluorophore with excellent photophysical properties and high ER accumulation ability, while several HTCF esters held a high potential to be good hCES2A substrates. Further investigations revealed that TP-HTCF (the tert-pentyl ester of HTCF) was an ideal substrate with ultrahigh sensitivity, excellent specificity, and a substantial signal-to-noise ratio. Upon the addition of hCES2A, TP-HTCF could be rapidly hydrolyzed to release HTCF, a chemically stable product that emitted bright fluorescent signals at around 670 nm. A TP-HTCF-based biochemical assay was then established for the high-throughput screening of potent and cell-active hCES2A inhibitors from an in-house compound library. Furthermore, TP-HTCF displayed high imaging resolution for imaging hCES2A in living cells as well as mouse liver slices and tumor-xenograft mice. Collectively, this study demonstrates a rational strategy for developing highly specific fluorogenic substrates for an ER-resident target enzyme, while TP-HTCF can act as a practical tool for sensing hCES2A in living systems.


Assuntos
Carboxilesterase , Corantes Fluorescentes , Humanos , Camundongos , Animais , Corantes Fluorescentes/química , Ensaios de Triagem em Larga Escala , Hidrólise , Ésteres
5.
Int J Biol Sci ; 19(11): 3360-3382, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37496997

RESUMO

Targeted therapies in cancer treatment can improve in vivo efficacy and reduce adverse effects by altering the tissue exposure of specific biomolecules. However, there are still large number of target proteins in cancer are still undruggable, owing to the following factors including (1) lack of ligand-binding pockets, (2) function based on protein-protein interactions (PPIs), (3) the highly specific conserved active sites among protein family members, and (4) the variability of tertiary docking structures. The current status of undruggable targets proteins such as KRAS, TP53, C-MYC, PTP, are carefully introduced in this review. Some novel techniques and drug designing strategies have been applicated for overcoming these undruggable proteins, and the most classic and well-known technology is proteolysis targeting chimeras (PROTACs). In this review, the novel drug development strategies including targeting protein degradation, targeting PPI, targeting intrinsically disordered regions, as well as targeting protein-DNA binding are described, and we also discuss the potential of these strategies for overcoming the undruggable targets. Besides, intelligence-assisted technologies like Alpha-Fold help us a lot to predict the protein structure, which is beneficial for drug development. The discovery of new targets and the development of drugs targeting them, especially those undruggable targets, remain a huge challenge. New drug development strategies, better extraction processes that do not disrupt protein-protein interactions, and more precise artificial intelligence technologies may provide significant assistance in overcoming these undruggable targets.


Assuntos
Inteligência Artificial , Neoplasias , Humanos , Proteínas/metabolismo , Proteólise , Neoplasias/tratamento farmacológico , Descoberta de Drogas
6.
J Med Chem ; 66(10): 6743-6755, 2023 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-37145039

RESUMO

Cytochrome P450 3A4 (CYP3A4) is a key xenobiotic-metabolizing enzyme-mediated drug metabolism and drug-drug interaction (DDI). Herein, an effective strategy was used to rationally construct a practical two-photon fluorogenic substrate for hCYP3A4. Following two-round structure-based substrate discovery and optimization, we have successfully constructed a hCYP3A4 fluorogenic substrate (F8) with desirable features, including high binding affinity, rapid response, excellent isoform specificity, and low cytotoxicity. Under physiological conditions, F8 is readily metabolized by hCYP3A4 to form a brightly fluorescent product (4-OH F8) that can be easily detected by various fluorescence devices. The practicality of F8 for real-time sensing and functional imaging of hCYP3A4 has been examined in tissue preparations, living cells, and organ slices. F8 also demonstrates good performance for high-throughput screening of hCYP3A4 inhibitors and assessing DDI potentials in vivo. Collectively, this study develops an advanced molecular tool for sensing CYP3A4 activities in biological systems, which strongly facilitates CYP3A4-associated fundamental and applied research studies.


Assuntos
Citocromo P-450 CYP3A , Corantes Fluorescentes , Citocromo P-450 CYP3A/metabolismo , Corantes Fluorescentes/farmacologia , Interações Medicamentosas
7.
Heliyon ; 9(3): e13840, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36879978

RESUMO

Background: Docetaxel (DCT) is widely used in clinical practice, but the drug resistance of breast cancer patients has become an important reason to limit its clinical efficacy. Chan'su is a commonly used traditional Chinese medicine for the treatment of breast cancer. Bufalin (BUF) is a bioactive polyhydroxy steroid extracted from chan'su and has strong antitumor activity, but there are few studies on reversing drug resistance in breast cancer. The aim of this study is to determine whether BUF can reverse the drug resistance to DCT and restore efficacy in breast cancer. Methodology: The reversal index of BUF was detected by Cell Counting Kit-8 (CCK-8) assays. The effects of BUF on enhancing the apoptosis of DCT were detected by flow cytometry and Western Blot (WB), and the main differential expression levels of sensitive and resistant strains were detected by high-throughput sequencing. Rhodamine 123 assay, WB and ATP Binding Cassette Subfamily B Member 1 (ABCB1) ATPase activity experiments were used to detect the effect of BUF on ABCB1. The nude mouse orthotopic model was constructed to investigate the reversal effect of BUF on DCT resistance in vivo. Results: With BUF intervention, the sensitivity of drug-resistant cell lines to DCT was increased. BUF can inhibit the expression of ABCB1 protein, increase the drug accumulation of DCT in drug-resistant strains, and reduce the ATPase activity of ABCB1. Animal experiments show that BUF can inhibit the growth of drug-resistant tumors in an orthotopic model of breast cancer and decrease the expression of ABCB1. Conclusion: BUF can reverse ABCB1-mediated docetaxel resistance in breast cancer.

8.
Phytomedicine ; 107: 154455, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36182797

RESUMO

BACKGROUND: Macrophages infiltration and activation play multiple roles in maintaining intestinal homeostasis and participate in the occurrence and development of UC. Thus, the restoration of immune balance can be achieved by targeting macrophage polarization. Previous studies have reported that TXYF could effectively ameliorate DSS-induced colitis. However, the underlying mechanisms of TXYF for DSS-induced colitis are still ill-defined. METHODOLOGY: This study was designed to explore the therapeutic effect of TXYF and its regulation in macrophages polarization during DSS-induced mice. In C75BL/6 mice, dextran sulfate sodium (DSS) was used to induce colitis and concomitantly TXYF was taken orally to evaluate its curative effect. In vitro experiment was implemented on BMDMs by lipopolysaccharide, IFN- and ATP. RESULTS: Here, we found that TXYF ameliorated clinical features in DSS-induced mice, decreased macrophages M1 polarization but remarkably increased M2 polarization. Mechanically, TXYF treatment effectively inhibited the activities of nuclear transcription factor NF-κB, which further contributed to the decrease of the inflammasome genes of NLRP3, limiting the activation of NLRP3 inflammasome in vivo and in vitro. CONCLUSION: Our findings demonstrated administration of TXYF can interfere with macrophage infiltration and polarization to improve the symptoms of acute colitis, by repressing NF-κB/NLRP3 signaling pathway activation. This enriches the mechanism and provides new prospect for TXYF in the treatment of colitis.


Assuntos
Colite , NF-kappa B , Trifosfato de Adenosina/metabolismo , Animais , Colite/induzido quimicamente , Colite/tratamento farmacológico , Sulfato de Dextrana/efeitos adversos , Medicamentos de Ervas Chinesas , Inflamassomos , Lipopolissacarídeos/farmacologia , Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Transdução de Sinais
9.
Phytomedicine ; 103: 154228, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35689898

RESUMO

BACKGROUND: Atopic dermatitis (AD), a common inflammatory skin disorder, severely affects the life quality of patients and renders heavy financial burden on patient's family. The Chinese medicine Viola yedoensis Makino formula (VYAC) has been widely used for treating various skin disorders. Previous studies have reported that VYAC is effective in relieving DNCB-induced AD and inflammation. However, the anti-inflammatory mechanism of VYAC is still ill-defined and poorly understood. This study aims to investigate the therapeutic effects of VYAC on DNCB-induced AD and to elucidate the underlying anti-inflammatory mechanisms. METHODOLOGY: VYAC were extracted with 70% ethanol and lyophilized for use. AD mice were established by DNCB. The therapeutic effects of VYAC were evaluated by oral administration VYAC (150, 300 and 600 mg/kg) daily in vivo. The histopathological and immunohistochemistry were used to analyze skin lesion and macrophages infiltration, RT-qPCR and Elisa were used to analyze the inflammatory factors in skin tissues and serum. To explore the underlying mechanism of VYAC against AD in vitro. RAW264.7 cells and bone-marrow-derived macrophages (BMDMs) were employed for macrophage polarization analysis. Flow cytometer, immunofluorescence and western blot were used to analyze M2 macrophages markers. STAT3 siRNA were transfected into both cells to validate the effects of VYAC-induced macrophages M2 polarization via JAK2/STAT3 signaling pathway. RESULTS: VYAC ameliorated skin lesion of DNCB-induced AD mice by decreased clinical scores and epidermal thickness, decreased the level of pro-inflammatory factors (IL-1ß, TNF-α and IL-18) and enhanced IL-10 anti-inflammatory factor level, inhibited macrophages infiltration and promoted M2 macrophages polarization in vivo. VYAC significantly promoted M2 macrophages polarization in vitro. It is observed that VYAC not only inhibited the phosphorylation of JAK2 and STAT3 in RAW264.7 cells and BMDMs, but also accelerated the translocation to the nucleus. What's more, VYAC reduced the polarization of M2 macrophage by activating JAK2/STAT3 signaling pathway was observed in both cells. CONCLUSIONS: Our findings demonstrate that VYAC significantly ameliorates skin lesion of DNCB-induced AD mice and reduces the levels of inflammatory factors by activating JAK2/STAT3 signaling pathway and promoting M2 macrophages polarization.


Assuntos
Dermatite Atópica , Medicamentos de Ervas Chinesas , Janus Quinase 2 , Macrófagos , Fator de Transcrição STAT3 , Viola , Animais , Anti-Inflamatórios/uso terapêutico , Polaridade Celular , Citocinas/metabolismo , Dermatite Atópica/induzido quimicamente , Dermatite Atópica/tratamento farmacológico , Dermatite Atópica/metabolismo , Dinitroclorobenzeno , Medicamentos de Ervas Chinesas/farmacologia , Janus Quinase 2/metabolismo , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Extratos Vegetais/farmacologia , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Viola/química
10.
J Ethnopharmacol ; 287: 114915, 2022 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-34954267

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Cucurbitacin B (CuB), extracted from muskmelon pedicel, is a widely available triterpenoid molecule that exerts influence on various biological activities. Modern pharmacological studies have found that cucurbitacin B has many kinds of pharmacological anti-tumor and anti-metastasis functions. AIM OF THE STUDY: To explore the mechanism of anti-tumor and anti-metastasis effect of cucurbitacin B. MATERIALS AND METHODS: The effect of cucurbitacin B on the growth of HCT116 and CT-26 was detected by CCK8; apoptosis was determined by flow cytometry and colony formation; the expression of apoptosis-related protein Bax, Bcl-2 and Cleaved-caspase-3 were examined by western Blot. To explore the underlying mechanism of cucurbitacin B against tumor, the Western blot, Immunofluorescence staining, Microscale Thermophoresis assays were used. Multiple molecular biology experiments were applied to validate the effect of polarization of cucurbitacin B-induced macrophages. The supernatant of Cucurbitacin B-induced macrophages and colon cells were co-cultured in vitro, and then transwell and wound healing assay were employed to the related phenotypes. C57BL/6 and BALB/c murine colon cancer model were also used to study the drug effects in vivo. RESULTS: Cucurbitacin B distinctly induced the apoptosis of CRC cells. It was observed that cucurbitacin B not only inhibited the phosphorylation of JAK2 and STAT3, but also the translocation from the cytosol to the nucleus. Meanwhile, we observed that cucurbitacin B is bound to STAT3. Further experimentation demonstrated that cucurbitacin B reduced the polarization of M2 macrophage by down-regulating JAK2/STAT3 signaling pathway. Cucurbitacin B-induced M2-like macrophages were found to diminish the migration of CRC cells. In vitro study suggested that cucurbitacin inhibited the CRC cells proliferation via JAK2/STAT3 and suppressed the cell migration by suppressing M2-like macrophages polarization. Consistent with in vitro results, the cucurbitacin B therapy significantly inhibited tumor growth and metastasis in mice. Moreover, in vivo the treatment with cucurbitacin B enhanced anti-tumor immunity by regulating M2-like macrophages and promoted the expression of CD4 and CD8 in tumor microenvironment. CONCLUSION: Our results proved that cucurbitacin B might be a potential candidate agent for adjuvant therapy in the process of CRC growth and metastasis.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Macrófagos/efeitos dos fármacos , Triterpenos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/patologia , Células HCT116 , Humanos , Janus Quinase 2/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Células RAW 264.7 , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células THP-1 , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Am J Cancer Res ; 11(9): 4308-4328, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34659889

RESUMO

Metastasis is the primary cause of death in lung cancer, one of the most prevalent and deadly neoplasms. The tumour-associated macrophages (TAMs) are crucial mediators to induce epithelial-mesenchymal transition (EMT) and promote lung metastasis via release of the cytokines. Matrine, a naturally occurring alkaloid, has been found with a variety of pharmacological effects, such as anti-cancer. In this study, an in vitro co-culture cell systems and a Lewis-bearing mouse model were employed to assay the potential effects of matrine on macrophages polarization, and its regulatory effects on EMT of Lewis lung cancer cells (LLCs). Our results clearly demonstrated that matrine inhibited M2-like RAW264.7 polarization, reducing the production of anti-inflammatory cytokines (IL-4, IL-10, and Arg-1), and M2 surface markers (CD206) were induced by LLCs via mTOR/PI3k/Akt signaling pathway, while it had no significant effect on M1 macrophages polarization. In vitro assays suggested that matrine partially blocked the metastasis of LLCs, and inhibited EMT induced by M2-like macrophages, which was evidenced by up-regulating the expression of E-cadherin and down-regulating the expression of N-cadherin, vimentin, and Snail. In vivo studies revealed that matrine decreased the ratio of CD206+/F4/80+, promoted the expression of CD4+ and CD8+ T cells, and inhibited the expression of Th2 in tumor and spleen tissues. Cell co-culture experiments revealed that Matrine promoted T-cell proliferation, which was impaired by tumour-derived CD11b+ myeloid cells. Collectively, our findings suggest that suppression of M2-like macrophages polarization of TAMs is a potential mechanism underlying the anti-metastasis effects of matrine in lung cancer.

12.
Basic Clin Pharmacol Toxicol ; 129(6): 437-449, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34478607

RESUMO

Methylophiopogonanone A (MOA) is an abundant homoisoflavonoid in the Chinese herb Ophiopogonis Radix. Recent investigations revealed that MOA inhibited several human cytochrome P450 enzymes (CYPs) and stimulated OATP1B1. However, the inhibitory effects of MOA on phase II drug-metabolizing enzymes, such as human UDP-glucuronosyltransferases (hUGTs), have not been well investigated. Herein, the inhibition potentials of MOA on hUGTs were assessed. The results clearly demonstrated that MOA dose-dependently inhibited all tested hUGTs including UGT1A1 (IC50 = 1.23 µM), one of the most important detoxification enzymes in humans. Further investigations showed that MOA strongly inhibited UGT1A1-catalysed NHPH-O-glucuronidation in a range of biological settings including hUGT1A1, human liver microsomes (HLM) and HeLa cells overexpressing UGT1A1. Inhibition kinetic analyses demonstrated that MOA competitively inhibited UGT1A1-catalysed NHPH-O-glucuronidation in both hUGT1A1 and HLM, with Ki values of 0.52 and 1.22 µM, respectively. Collectively, our findings expanded knowledge of the interactions between MOA and human drug-metabolizing enzymes, which would be very helpful for guiding the use of MOA-related herbal products in clinical settings.


Assuntos
Benzodioxóis/farmacologia , Inibidores Enzimáticos/farmacologia , Glucuronosiltransferase/antagonistas & inibidores , Interações Ervas-Drogas , Isoflavonas/farmacologia , Benzodioxóis/administração & dosagem , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/administração & dosagem , Células HeLa , Humanos , Concentração Inibidora 50 , Isoflavonas/administração & dosagem , Microssomos Hepáticos/enzimologia
13.
Front Oncol ; 11: 634851, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33718223

RESUMO

Lung cancer is one of the most common and lethal neoplasms for which very few efficacious treatments are currently available. M1-like polarised tumour-associated macrophages (TAMs) are key mediators to modulate the tumour microenvironment, which play a key role in inhibiting cancer cell growth. Sophoridine, a naturally occurring alkaloid, exerts multiple pharmacological activities including anti-tumour and anti-inflammatory activities, but it has not been characterised as a regulator of tumour microenvironment towards NSCLC. Herein, the regulatory effects of sophoridine on the polarisation of THP-1 cells into TAMs and the anti-tumour effects of sophoridine-stimulated M1 polarised macrophages towards lung cancer cells were carefully investigated both in vitro and in vivo. The results showed that sophoridine could significantly promote M1 polarisation of RAW264.7 and THP-1-derived macrophages, leading to increased expression of pro-inflammatory cytokines and the M1 surface markers CD86 via activating MAPKs signaling pathway. Further investigations showed that sophoridine-stimulated RAW264.7 and THP-1-derived M1 macrophages effectively induced cell apoptosis as well as inhibited the cell colony formation and cell proliferation in both H460 and Lewis lung cancer cells. In Lewis-bearing mice model, sophoridine (15 or 25 mg/kg) significantly inhibited the tumour growth and up-regulated the expression of CD86/F4/80 in tumour tissues. Collectively, the findings clearly demonstrate that sophoridine promoted M1-like polarisation in vitro and in vivo, suggesting that sophoridine held a great therapeutic potential for treating lung cancer.

14.
Biomed Pharmacother ; 138: 111449, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33706133

RESUMO

Qingfei oral liquid (QF) is a traditional Chinese medicine that has been used to treat patients with viral pneumonia and asthma for decades. Our previous study revealed that QF prevents airway inflammation and reduces airway hyperresponsiveness (AHR) in respiratory syncytial virus (RSV)-infected asthmatic mice. RSV infection can exacerbate asthma in pediatric patients and induce autophagy, which leads to the promotion of inflammatory cytokine production in the pathology of this disease. The effect of QF on regulating autophagy in RSV-infected asthma patients has not been fully elucidated. In this study, we identified compounds of QF by HPLC-DAD-Q-TOF-MS/MS. The RSV infected OVA challenged mice, we evaluated the RSV-infected asthma model. We found that treatment with QF alleviated airway inflammation and mitigated airway AHR in RSV-infected asthmatic mice. In addition, we found that QF inhibited autophagosome formation and the expression of LC3 protein by using electron and laser confocal microscopy, respectively, to assess RSV-infected asthmatic mice lung tissues. Furthermore, QF was found to reduce the quantity of autophagy and its related proteins LC3B (light chain 3B), Beclin-1, p62 and Atg5 (autophagy-related gene 5) and downstream inflammatory cytokines TNF-α, IL-4, IL-6, and IL-13 via an action in mTOR-dependent signaling in vivo and in vitro. These findings suggest that QF can alleviate the inflammation caused by RSV infection in asthmatic mice, and its mechanism may be involved in the regulation of autophagy via the mTOR signaling pathway.


Assuntos
Asma/metabolismo , Autofagia/efeitos dos fármacos , Medicamentos de Ervas Chinesas/administração & dosagem , Mediadores da Inflamação/metabolismo , Infecções por Vírus Respiratório Sincicial/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Administração Oral , Animais , Asma/tratamento farmacológico , Asma/imunologia , Autofagia/fisiologia , Linhagem Celular , Células Cultivadas , Humanos , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Infecções por Vírus Respiratório Sincicial/imunologia , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/imunologia
15.
Front Pharmacol ; 12: 747992, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35115921

RESUMO

Bufalin (Buf), an active ingredient of the traditional Chinese medicine Chansu, is known to have anticancer effects for breast cancer. However, its poor solubility, high toxicity, and extensive side effects limit its use. Metal-organic frameworks (MOFs) are a class of promising drug delivery systems known for their high porosity. Here, we designed and constructed pH-sensitive and redox-responsive folic acid-modified MOFs as drug carriers of Buf (FA-MOF/Buf). Moreover, the anticancer activity of nanomedicines was also explored in vitro and in vivo. Compared to free Buf, the FA-MOF/Buf nanoparticles demonstrated improved water solubility and stability, higher intracellular uptake, and enhanced cytotoxicity in breast cancer cells in vitro. Furthermore, it displayed improved accumulation in the tumor site, enhanced anticancer activity, and reduced side effects in vivo. Our results demonstrated that FA-MOF could be developed as a potential delivery system for Buf to improve its antitumor activity for breast cancer treatment.

16.
Biomed Pharmacother ; 128: 110340, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32521453

RESUMO

Pediatric asthma is exacerbated by Respiratory Syncytial Virus (RSV) infection, and Transient Receptor Potential Vanilloid 1 (TRPV1) promotes production of inflammatory cytokines and mucus hypersecretion in the pathology of this disease. Our previous research revealed that Qingfei oral liquid (QF) inhibited airway inflammation and mucus hypersecretion in RSV-infected asthmatic mice models and that this may be associated with the TRPV1-regulation of NF-κB and Mucin 5AC (MUC5AC) expression, but the exact mechanism is unknown. In the present study, LC-MS was used for analyzing the chemicals in QF, ovalbumin (OVA)-induced asthmatic mice inhaled RSV three consecutive times to create an RSV-infected asthmatic model. We found treatment from QF alleviated airway hyperresponsiveness (AHR) and reduced congestion, edema, and infiltration of inflammatory cells into pulmonary tissues. Additionally, QF was found to decrease expression of NF-κB and its downstream inflammatory cytokines IL-1ß, IL-4, IL-5, and IL-13, as well as a decrease in MUC5AC and pro-inflammatory cytokines in PKC via a reduction in Protein Kinase C-dependent signaling. These findings suggest that QF can alleviate AHR and mucus hypersecretion caused by RSV infection in asthmatic mice, and its mechanism may be associated with the regulation of the TRPV1 signaling pathway.


Assuntos
Asma/tratamento farmacológico , Hiper-Reatividade Brônquica/tratamento farmacológico , Broncoconstrição/efeitos dos fármacos , Medicamentos de Ervas Chinesas/administração & dosagem , Pulmão/efeitos dos fármacos , Mucina-5AC/metabolismo , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Canais de Cátion TRPV/antagonistas & inibidores , Administração Oral , Animais , Asma/metabolismo , Asma/fisiopatologia , Asma/virologia , Hiper-Reatividade Brônquica/metabolismo , Hiper-Reatividade Brônquica/fisiopatologia , Hiper-Reatividade Brônquica/virologia , Citocinas/metabolismo , Modelos Animais de Doenças , Mediadores da Inflamação/metabolismo , Pulmão/metabolismo , Pulmão/fisiopatologia , Pulmão/virologia , Masculino , Camundongos Endogâmicos BALB C , NF-kappa B/metabolismo , Proteína Quinase C/metabolismo , Infecções por Vírus Respiratório Sincicial/metabolismo , Infecções por Vírus Respiratório Sincicial/fisiopatologia , Infecções por Vírus Respiratório Sincicial/virologia , Via Secretória , Transdução de Sinais , Canais de Cátion TRPV/metabolismo
17.
Inflammation ; 43(4): 1423-1435, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32388657

RESUMO

Ulcerative colitis (UC) is a chronic and relapsing inflammatory disorder of the gastrointestinal tract, characterized by diarrhea, hematochezia, abdominal distension, and abdominal pain. The perpetuation of inflammation and the impairment of the intestinal barrier are part of the main courses of UC, responsible for the deteriorating inflammatory condition. Patchouli alcohol (PA), extracted from Pogostemon cablin Benth., is employed to treat both inflammation and intestinal barrier damage. Its curative effect on UC was testified firstly by TNBS-induced UC, a chemically induced colitis, and further tested by DSS-induced UC, an acute attack stage of UC in which the clinical course of human UC occurs frequently. PA reduced the levels of TNF-α, IFN-γ, IL-1ß, IL-6, and IL-17 in serum and decreased the mRNA expression of pro-inflammatory cytokines (e.g., iNOS, COX-2, TNF-α, IL-1ß, and IL-6). Concurrently, PA upregulated the expression of tight junction protein (e.g., ZO-1, ZO-2, claudin-1, and occludin) and the mRNA of mucin-1 and mucin-2 in both animal models. Further, PA ameliorated both histological damage and clinical parameters. Thus, PA could credibly reduce the expression of pro-inflammatory cytokines, protect the integrity of intestinal epithelial barrier, and repair the macroscopic colon lesions in both colitis models.


Assuntos
Colite Ulcerativa/patologia , Colite Ulcerativa/prevenção & controle , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/patologia , Sesquiterpenos/uso terapêutico , Animais , Colite Ulcerativa/induzido quimicamente , Sulfato de Dextrana/toxicidade , Feminino , Inflamação/induzido quimicamente , Inflamação/patologia , Inflamação/prevenção & controle , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Ratos , Ratos Sprague-Dawley , Sesquiterpenos/farmacologia
18.
Int J Pharm ; 580: 119123, 2020 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-32035258

RESUMO

The development of small molecule anticancer drugs, with low water solubility and high toxicity, into polymeric prodrugs has developed into a promising strategy in clinical application. In this study, we synthesized a novel G3-C12-mediated esterase-sensitive tumor-targeting polymeric prodrug of camptothecin (CPT), P(OEGMA-co-CPT-co-G3-C12), and explored its anticancer activity against androgen-independent prostate cancer in vitro and in vivo. Compared to free CPT, the multifunctional polymeric prodrug demonstrated improved water solubility and stability, higher intracellular uptake, and enhanced cytotoxicity in DU145 cells in vitro. Furthermore, it displayed an improved accumulation in the tumor and an enhanced anticancer activity in vivo. Hence, P(OEGMA-co-CPT-co-G3-C12) could be a promising drug in the treatment of androgen-independent prostate cancer.


Assuntos
Proteínas Sanguíneas/metabolismo , Camptotecina/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Desenvolvimento de Medicamentos/métodos , Galectinas/metabolismo , Pró-Fármacos/metabolismo , Neoplasias de Próstata Resistentes à Castração/metabolismo , Animais , Camptotecina/administração & dosagem , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Relação Dose-Resposta a Droga , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Polímeros/administração & dosagem , Polímeros/metabolismo , Pró-Fármacos/administração & dosagem , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/patologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
19.
J Control Release ; 320: 363-380, 2020 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-32001299

RESUMO

Macrophages, an important component of the innate immune response, are a key regulator of intestinal microenvironment homeostasis. These cells essentially contribute to chronic inflammatory diseases due to their strong plasticity. As is known, ulcerative colitis (UC), a chronic inflammatory disease, is closely related to immune dysfunction. A growing body of evidence suggests that the macrophage is a promising drug target for modulating the intestinal immune systems and regulating the inflammatory microenvironment, thus alleviating the inflammatory responses in UC. The macrophage-based therapy strategies for UC are still at an emerging stage. The advanced drug delivery systems can improve the macrophage-based therapy. This article will review the molecular mechanisms related to macrophage polarization and the interactions between signaling pathways that regulate the pathogenesis of UC and summarize the macrophage-based nanotherapeutic strategies in UC.


Assuntos
Colite Ulcerativa , Colite Ulcerativa/tratamento farmacológico , Humanos , Intestinos , Ativação de Macrófagos , Macrófagos , Transdução de Sinais
20.
Zhongguo Zhong Yao Za Zhi ; 44(14): 3042-3048, 2019 Jul.
Artigo em Chinês | MEDLINE | ID: mdl-31602851

RESUMO

To establish the spectrum-effect relationship between HPLC fingerprint and free radicals activity scavenging in Guizhi Shaoyao Zhimu Decoction( GSZD),and provide a basis for the quality evaluation and modernization of classical prescriptions. Shimadsu GL-science C18 column( 4. 6 mm×250 mm,5 µm) was used with acetonitrile-0. 1% formic acid solution as the mobile phase for gradient elution. The detective wave length was 254 nm; the column temperature was set at 32 ℃; the injection volume was 20 µL; and the flow rate was 1. 0 m L·min-1.10 batches of primary standard samples of GSZD were detected,and their HPLC fingerprint was established by using the similarity evaluation system for chromatographic fingerprint of traditional Chinese medicine( TCM). The activity of scavenging free radicals was studied by 1,1-diphenyl-2-trinitrophenylhydrazine( DPPH) method,and the spectrum-effect relationship was studied by Pearson bivariate correlation analysis. The common mode of GSZD fingerprints was established,and 26 common peaks were marked,with similarities ranging from 0. 929 to 0. 998. Eight of the chromatographic peaks were identified by using the control comparison method: gallic acid,mangiferin,paeoniflorin,glycyrrhizin,asparagus,5-O-methylvisamicin,cinnamic acid,and ammonium glycyrrhetate. Among them,the content changes of No. 14( paeoniside),20,12( mangiferin),13 and 23( cinnamic acid) common peaks were negatively correlated with free radical scavenging activity. The fingerprint showed high precision,repeatability and stability,and the common peaks were well separated,so it can be used for the quality evaluation of GSZD,and could provide reference for further studies on the material basis of GSZD.


Assuntos
Cinnamomum aromaticum/química , Medicamentos de Ervas Chinesas/química , Sequestradores de Radicais Livres/química , Cromatografia Líquida de Alta Pressão , Medicina Tradicional Chinesa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...