Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Phytomedicine ; 126: 155348, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38335913

RESUMO

BACKGROUND: (-)-Asarinin (Asarinin) is the primary component in the extract of the herb Asarum sieboldii Miq. It possesses various functions, including pain relief, anti-viral and anti-tuberculous bacilli effects, and inhibition of tumor growth. Gastric precancerous lesion (GPL) is a common but potentially carcinogenic chronic gastrointestinal disease, and its progression can lead to gastric dysfunction and cancer development. However, the protective effects of asarinin against GPL and the underlying mechanisms remain unexplored. METHODS: A premalignant cell model (methylnitronitrosoguanidine-induced malignant transformation of human gastric epithelial cell strain, MC cells) and a GPL animal model were established and then were treated with asarinin. The cytotoxic effect of asarinin was assessed using a CCK8 assay. Detection of intracellular reactive oxygen species (ROS) using DCFH-DA. Apoptosis in MC cells was evaluated using an annexin V-FITC/PI assay. We performed western blot analysis and immunohistochemistry (IHC) to analyze relevant markers, investigating the in vitro and in vivo therapeutic effects of asarinin on GPL and its intrinsic mechanisms. RESULTS: Our findings showed that asarinin inhibited MC cell proliferation, enhanced intracellular ROS levels, and induced cell apoptosis. Further investigations revealed that the pharmacological effects of asarinin on MC cells were blocked by the ROS scavenger N-acetylcysteine. IHC revealed a significant upregulation of phospho-signal transducer and activator of transcription 3 (p-STAT3) protein expression in human GPL tissues. In vitro, asarinin exerted its pro-apoptotic effects in MC cells by modulating the STAT3 signaling pathway. Agonists of STAT3 were able to abolish the effects of asarinin on MC cells. In vivo, asarinin induced ROS accumulation and inhibited the STAT3 pathway in gastric mucosa of mice, thereby halting and even reversing the development of GPL. CONCLUSION: Asarinin induces apoptosis and delays the progression of GPL by promoting mitochondrial ROS production, decreasing mitochondrial membrane potential (MMP), and inhibiting the STAT3 pathway.


Assuntos
Dioxóis , Lignanas , Lesões Pré-Cancerosas , Humanos , Camundongos , Animais , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Lignanas/farmacologia , Proliferação de Células , Lesões Pré-Cancerosas/induzido quimicamente , Lesões Pré-Cancerosas/tratamento farmacológico , Lesões Pré-Cancerosas/patologia , Apoptose , Fator de Transcrição STAT3/metabolismo , Linhagem Celular Tumoral
2.
J Ethnopharmacol ; 324: 117745, 2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38228231

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Jin-Gui-Shen-Qi Wan (JGSQW) is a traditional Chinese medicine formula that has been traditionally used to alleviate urinary system ailments such as frequent urination and polyuria. Clinical studies have indicated that when combined with hypoglycaemic drugs, JGSQW exhibits a synergistic effect and can improve diabetic nephropathy (DN), yet its underlying mechanism and targets remain unclear. AIM OF THE STUDY: This study aims to investigate the therapeutic efficacy of JGSQW and its underlying mechanisms using a DN db/db mouse model. MATERIALS AND METHODS: Ultrahigh-performance liquid chromatography coupled with mass spectrometry was utilized to analyse the primary active compounds, blood levels, and pharmacokinetics of JGSQW. Additionally, the therapeutic effects of JGSQW and metformin on blood glucose levels, lipid levels, renal function, and renal pathology in diabetic nephropathy mice were investigated using a db/db mouse model. Proteomic analysis was carried out to identify the primary target of JGSQW in treating DN. The mechanism of action was verified by western blotting, immunohistochemistry, and immunofluorescence. Then, molecular docking and molecular dynamics, transfection, drug affinity responsive target stability (DARTS) assay and cell thermal migration assay (CETSA) further validated the targeted binding effect. RESULTS: JGSQW combined with metformin significantly improved the blood glucose levels, blood lipids, renal function, and renal pathology of DN mice. JGSQW mainly exerted its therapeutic effect on DN by targeting major histocompatibility complex class II (MHC class II) molecules. Immunohistochemistry results showed that JGSQW inhibited the expression of collagen I, fibronectin, and alpha smooth muscle actin (α-SMA) expression. Immunofluorescence and Western blot results showed that JGSQW inhibited the expression of H2-Ab1 and H2-Aa, which are MHC class II molecules, thereby suppressing CD4+ T-cell infiltration and improving diabetic kidney fibrosis. The binding ability of paeoniflorin to H2-Aa was predicted and verified by molecular, DARTS, and CETSA assays. Treatment with 80 µM paeoniflorin effectively alleviated high glucose-induced injury in the MPC-5 injury model. H2-Aa was overexpressed at this model concentration, and Western blotting further confirmed that paeoniflorin reduced glomerular podocyte fibrosis by regulating H2-Aa. CONCLUSIONS: JGSQW combined with metformin may have a synergistic effect to alleviates renal fibrosis in diabetic nephropathy by downregulating immune complex MHC class II molecules and attenuating the antigen presentation effect of MHC class II on CD4.


Assuntos
Diabetes Mellitus , Nefropatias Diabéticas , Glucosídeos , Metformina , Monoterpenos , Camundongos , Animais , Nefropatias Diabéticas/patologia , Glicemia , Simulação de Acoplamento Molecular , Proteômica , Transdução de Sinais , Fibrose , Antígenos de Histocompatibilidade Classe II/farmacologia , Antígenos de Histocompatibilidade Classe II/uso terapêutico , Metformina/farmacologia , Metformina/uso terapêutico
3.
Chin Med ; 18(1): 130, 2023 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-37828620

RESUMO

BACKGROUND: Jin-Gui-Shen-Qi Wan (JGSQ) has been used in China for thousands of years to treat various ailments, including frequent urination, blurred vision, and soreness in the waist and knees. It has traditional therapeutic advantages in improving eye diseases. AIM OF THE STUDY: Clinical studies have confirmed the therapeutic efficacy of JGSQ in improving diabetes and vision; however, its efficacy and pharmacological effects in treating diabetic retinopathy (DR) remain unclear. Therefore, the aim of this study was to investigate the specific pharmacological effects and potential mechanisms of JGSQ in improving DR through a db/db model. MATERIALS AND METHODS: db/db mice were given three different doses of orally administered JGSQ and metformin for 8 weeks, and then PAS staining of the retinal vascular network patch, transmission electron microscopy, H&E staining, and TUNEL staining were performed to determine the potential role of JGSQ in improving DR-induced neuronal cell apoptosis. Furthermore, network pharmacology analysis and molecular docking were carried out to identify the main potential targets of JGSQ, and the efficacy of JGSQ in improving DR was evaluated through western blotting and immunofluorescence staining, revealing its mechanism of action. RESULTS: According to the results from H&E, TUNEL, and PAS staining of the retinal vascular network patch and transmission electron microscopy, JGSQ does not have an advantage in improving the abnormal morphology of vascular endothelial cells, but it has a significant effect on protecting retinal ganglion cells from apoptosis. Through network pharmacology and molecular docking, AKT, GAPDH, TNF, TP53, and IL-6 were identified as the main core targets of JGSQ. Subsequently, through western blot and immunofluorescence staining, it was found that JGSQ can inhibit HIF-1α, promote p-AKT expression, and inhibit TP53 expression. At the same time, inhibiting the release of inflammatory factors protects retinal ganglion cells and improves apoptosis in DR. CONCLUSION: These results indicated that in the db/db DR mouse model, JGSQ can inhibit the expression of inflammatory cytokines and protect retinal ganglion cells from apoptosis, possibly by modulating the Akt/HIF-1α pathway.

4.
MedComm (2020) ; 4(5): e359, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37692109

RESUMO

Purines and purinergic receptors are widely distributed throughout the human body. Purine molecules within cells play crucial roles in regulating energy metabolism and other cellular processes, while extracellular purines transmit signals through specific purinergic receptors. The ubiquitous purinergic signaling maintains normal neural excitability, digestion and absorption, respiratory movement, and other complex physiological activities, and participates in cell proliferation, differentiation, migration, and death. Pathological dysregulation of purinergic signaling can result in the development of various diseases, including neurodegeneration, inflammatory reactions, and malignant tumors. The dysregulation or dysfunction of purines and purinergic receptors has been demonstrated to be closely associated with tumor progression. Compared with other subtypes of purinergic receptors, the P2X7 receptor (P2X7R) exhibits distinct characteristics (i.e., a low affinity for ATP, dual functionality upon activation, the mediation of ion channels, and nonselective pores formation) and is considered a promising target for antitumor therapy, particularly in patients with poor response to immunotherapy This review summarizes the physiological and pathological significance of purinergic signaling and purinergic receptors, analyzes their complex relationship with tumors, and proposes potential antitumor immunotherapy strategies from tumor P2X7R inhibition, tumor P2X7R overactivation, and host P2X7R activation. This review provides a reference for clinical immunotherapy and mechanism investigation.

5.
Int Immunopharmacol ; 124(Pt A): 110902, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37699302

RESUMO

BACKGROUND: Gastric precancerous lesions (GPLs) are omens for gastric cancer (GC), which developing with a series of pathological changes of gastric mucosa. Reversing epithelial-mesenchymal transition (EMT) in gastric mucosa is the main approach to restrain GPLs from evolving into cancer. Tanshinone I (Tan-I), the active ingredients of traditional Chinese herb Salvia miltiorrhiza, has exhibited anticancer effect. PURPOSE: To investigate the effect and mechanism of Tan-I in intervening GPLs, and provide a new therapeutic strategy for prevention of GC. METHODS: Gastric mucosal epithelial cells were treated with the N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) to construct MNNG-induced cell (MC cell) of gastric mucosa that undergoing EMT process. Then, this study explored the effect and mechanism of Tan-I in vitro. Subsequently, this study constructed GPL mice to clarify the exact efficacy and mechanism of Tan-I on GPLs. RESULTS: Tan-I inhibited MC cell proliferation, invasion and migration. Simultaneously, the aberrant expression of E-cadherin and N-cadherin were reversed. Tan-I attenuated inflammation by reducing the release of nitric oxide, TNFα and IL-1ß. Tan-I reversed the EMT and inflammatory processes by regulating p38 and STAT3. CONCLUSION: This study showed that Tan-I inhibited the progression of GPLs by reversing the EMT process and reducing inflammation by restraining the p38/STAT3 signaling pathway.

6.
Front Pharmacol ; 14: 1186712, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37560476

RESUMO

Extracellular matrix (ECM) plays a pivotal and dynamic role in the construction of tumor microenvironment (TME), becoming the focus in cancer research and treatment. Multiple cell signaling in ECM remodeling contribute to uncontrolled proliferation, metastasis, immune evasion and drug resistance of cancer. Targeting trilogy of ECM remodeling could be a new strategy during the early-, middle-, advanced-stages of cancer and overcoming drug resistance. Currently nearly 60% of the alternative anticancer drugs are derived from natural products or active ingredients or structural analogs isolated from plants. According to the characteristics of ECM, this manuscript proposes three phases of whole-process management of cancer, including prevention of cancer development in the early stage of cancer (Phase I); prevent the metastasis of tumor in the middle stage of cancer (Phase II); provide a novel method in the use of immunotherapy for advanced cancer (Phase III), and present novel insights on the contribution of natural products use as innovative strategies to exert anticancer effects by targeting components in ECM. Herein, we focus on trilogy of ECM remodeling and the interaction among ECM, cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs), and sort out the intervention effects of natural products on the ECM and related targets in the tumor progression, provide a reference for the development of new drugs against tumor metastasis and recurrence.

7.
Redox Rep ; 28(1): 2243423, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37565601

RESUMO

BACKGROUND: Acute lung injury (ALI) is a severe and often fatal pulmonary disease. Current treatments for ALI and acute respiratory distress syndrome (ARDS) are limited. Natural product metabolites have shown promise as therapeutic alternatives. However, the effects of Licochalcone B (LCB) on ALI are largely unknown. METHODS: We investigated the effects of LCB on lipopolysaccharide-challenged mice and human pulmonary microvascular endothelial cells. Cell viability, apoptosis, and ROS production were assessed. Lung tissue histopathology and oxidative stress and inflammation markers were evaluated. Protein expression levels were measured. RESULTS: LCB had no cytotoxic effects on cells and increased cell viability. It reduced apoptosis and ROS levels in cells. In mice with ALI, LCB decreased lung tissue weight and improved oxidative stress and inflammation markers. It also enhanced expression levels of Nrf2, HO-1, and NQO1 while reducing Keap1. CONCLUSION: LCB protects against LPS-induced acute lung injury in cells and mice. The Keap1/Nrf2 pathway may be involved in its protective effects. LCB shows potential as a strategy to alleviate ALI caused by LPS.


Assuntos
Lesão Pulmonar Aguda , Animais , Humanos , Camundongos , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/tratamento farmacológico , Células Endoteliais/metabolismo , Inflamação/patologia , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Lipopolissacarídeos/toxicidade , Pulmão/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo
8.
Phytother Res ; 37(10): 4674-4689, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37402476

RESUMO

Chelerythrine chloride (CHE) is a benzodiazepine alkaloid derived from natural herbs with significant anti-tumor and anti-inflammatory activities. However, the exact role and underlying mechanisms of CHE in colorectal cancer (CRC) remain unclear. Therefore, this study is aimed to investigate the influence of CHE on the progression of CRC. Cell Counting Kit-8 assay (CCK-8), transwell, apoptosis rate, cell cycle distribution, reactive oxygen species (ROS), and colony formation determined the anti-proliferative activity of CHE in CRC cell lines. Transcriptome sequencing and western blot were used to explore the mechanism. Finally, H&E staining, Ki67, TUNEL, and immunofluorescence were conducted to verify the anti-CRC activity and potential mechanisms of CHE in vivo. CHE had a prominent inhibitory effect on the proliferation of CRC cells. CHE induces G1 and S phase arrest and induces cell apoptosis by ROS accumulation. Cancer-associated fibroblasts (CAFs) play a key role in CRC metastasis. Then, this study found that CHE regulates WNT10B/ß-catenin and TGFß2/Smad2/3 axis, thereby decreasing the expression of α-SMA, which is a maker of CAFs. Taken together, CHE is a candidate drug and a potent compound for metastatic CRC, which can intervene CAFs in a dual pathway to effectively inhibit the invasion and migration of cancer cells, which can provide a new choice for future clinical treatment.

9.
Phytomedicine ; 115: 154828, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37116386

RESUMO

BACKGROUND: Retinitis pigmentosa (RP) refers to a group of progressive photoreceptor degenerative diseases. The activation of microglia has been reported to play an important role in the photoreceptor degeneration in RP retinal. Bujing Yishi tablets (BJYS), a Chinese herbal medicine, has been used to treat retinal diseases in China with desirable effect in improving visual function. However, the mechanisms underlying the efficacy of BJYS treatment in RP are not yet fully understood. PURPOSE: Based on the preliminary experiments, this study aimed to investigate the therapeutic mechanism involved in treating N-Methyl-N-Nitrosourea (MNU)-induced retinal degeneration of RP with BJYS. METHODS: To explore the efficacy of BJYS, a rat experimental RP model was established through intraperitoneal injection of MNU (50 mg/kg). Two experiment was carried out. After the treatment, we conducted H&E, TUNEL, retinal cytokine levels and IBA-1 expression in microglia to confirm the impact on RP model. The specific mechanism of action of BJYS tablet was assessed by western blot, real-time polymerase chain reaction (RT-PCR), and immunofluorescence to determine the mRNA and protein expression levels involved in clarifying the effectiveness of BJYS exerted through P2X7R/CX3CL1/CX3CR1 pathway. RESULTS: Significant alleviation of retinal morphological structure and photoreceptor degeneration by BJYS treatment was observed in the retinal of MNU-induced RP rats, BJYS prevented the reduction of ONL thickness and decreased the level of apoptotic cells in ONL. It also inhibited microglia overactivation and reduced retinal levels of IL-1ß, IL-6, TNF-α. In addition, BJYS decreased the protein expression and mRNA expression of P2X7, CX3CL1 and CX3CR1 and reduced the phosphorylation of p38 MAPK. CONCLUSION: In summary, this study suggested that BJYS treatment could alleviate photoreceptors degeneration of RP by inhibiting microglia overactivation and inflammation through the P2X7R/CX3CL1/CX3CR1 pathway. These effects suggest that BJYS tablets may serve as a promising oral therapeutic agent for RP.


Assuntos
Degeneração Retiniana , Retinite Pigmentosa , Ratos , Animais , Retinite Pigmentosa/tratamento farmacológico , Retinite Pigmentosa/metabolismo , Células Fotorreceptoras/metabolismo , Retina , Degeneração Retiniana/induzido quimicamente , Morte Celular , Compostos de Nitrosoureia/efeitos adversos , Compostos de Nitrosoureia/metabolismo , Apoptose , Modelos Animais de Doenças , Quimiocina CX3CL1/efeitos adversos , Quimiocina CX3CL1/metabolismo , Receptor 1 de Quimiocina CX3C/metabolismo
10.
Phytomedicine ; 110: 154608, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36586205

RESUMO

BACKGROUND: Mitochondria are the energy factories of cells with the ability to modulate the cell cycle, cellular differentiation, signal transduction, growth, and apoptosis. Existing drugs targeting mitochondria in cancer treatment have disadvantages of drug resistance and side effects. Phytochemicals, which are widely found in plants, are bioactive compounds that could facilitate the development of new drugs for gastric cancer. Studies have shown that some phytochemicals can suppress the development of gastric cancer. METHODS: We searched for data from PubMed, China National Knowledge Infrastructure, Web of Science, and Embase databases from initial establishment to December 2021 to review the mechanism by which phytochemicals suppress gastric cancer cell growth by modulating mitochondrial function. Phytochemicals were classified and summarized by their mechanisms of action. RESULTS: Phytochemicals can interfere with mitochondria through several mechanisms to reach the goal of promoting apoptosis in gastric cancer cells. Some phytochemicals, e.g., daidzein and tetrandrine promoted cytochrome c spillover into the cytoplasm by modulating the members of the B-cell lymphoma-2 protein family and induced apoptotic body activity by activating the caspase protein family. Phytochemicals (e.g., celastrol and shikonin) could promote the accumulation of reactive oxygen species and reduce the mitochondrial membrane potential. Several phytochemicals (e.g., berberine and oleanolic acid) activated mitochondrial apoptotic submission via the phosphatidylinositol-3-kinase/Akt signaling pathway, thereby triggering apoptosis in gastric cancer cells. Several well-known phytochemicals that target mitochondria, including berberine, ginsenoside, and baicalein, showed the advantages of multiple targets, high efficacy, and fewer side effects. CONCLUSIONS: Phytochemicals could target the mitochondria in the treatment of gastric cancer, providing potential directions and evidence for clinical translation. Drug discovery focused on phytochemicals has great potential to break barriers in cancer treatment.


Assuntos
Berberina , Neoplasias Gástricas , Humanos , Berberina/farmacologia , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/metabolismo , Mitocôndrias , Transdução de Sinais , Apoptose , Compostos Fitoquímicos/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo , Linhagem Celular Tumoral
11.
J Ethnopharmacol ; 303: 116025, 2023 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-36496042

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Berberis amurensis Rupr. is used to treat cancer as a traditional herbal medicine. Berbamine (BBM) is a natural bisbenzylisoquinoline alkaloid extracted from Berberis amurensis which possesses multiple pharmacological activity including anticancer. AIM OF THE STUDY: To investigate the influence of BBM on the progression of colorectal cancer (CRC) and further explore the underlying mechanism of BBM based on the RTKs/Akt signaling pathway. MATERIALS AND METHODS: In vitro, cell viability and colony formation were conducted to detect BBM inhibitory of CRC cell lines. Transwell was detected the ability of migration and invasion by BBM. Apoptosis detection assay, cell cycle assay and the measurement of ROS were detected to confirm the inductive effect of cell apoptosis. RT-qPCR and Western blot to clarify the specific mechanism of anticancer. Finally, we conducted HE staining, Ki67, Tunnel and immunochemistry were confirmed the anti-colorectal cancer activity of BBM from vivo study. RESULTS: We found that BBM could inhibit CRC cell lines growth. Moreover, BBM presented an inhibitory effect the ability of migration and invasion in CRC cells. Furthermore, the occurrence of apoptosis was involved in the anti-colorectal cancer role of BBM. BBM also triggered ROS accumulation in CRC cells that might be a key factor for the inductive effect of BBM in cell apoptosis. Cell cycle assay revealed that BBM induced the arrest of G1-S phase and increased the p21 levels but decreased CyclinE1, CyclinE2, CDK6, CyclinD1. RT-qPCR manifested that the down-regulation effect of BBM on AKT1, EGFR, PDGFRα and FGFR4 genes. The results also showed that BBM could decreased the expression levels of phosphor-AKT, PDGFRα, PDGFRß, EGFR, FGFR3 and FGFR4 which belong to RTKs family. Consistently, BBM remarkably suppressed tumor xenograft growth in nude mice. CONCLUSION: Taken together, all the results as presented above suggest that BBM as a novel multitargeted receptor tyrosine kinase inhibitor plays a crucial role in the inhibitory effect of CRC and may be a promising therapeutic agent for the CRC in clinic.


Assuntos
Benzilisoquinolinas , Neoplasias Colorretais , Camundongos , Animais , Humanos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Camundongos Nus , Espécies Reativas de Oxigênio , Receptor alfa de Fator de Crescimento Derivado de Plaquetas , Neoplasias Colorretais/patologia , Benzilisoquinolinas/farmacologia , Benzilisoquinolinas/uso terapêutico , Apoptose , Receptores ErbB/metabolismo , Proliferação de Células , Linhagem Celular Tumoral , Movimento Celular
12.
J Ethnopharmacol ; 302(Pt A): 115885, 2023 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-36328204

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Gallic acid (GA) is a natural polyphenolic compound derived from Rhus chinensis Mill. with a variety of biological activities such as astringent sweat, cough, dysentery, hemostasis, and detoxification, and is widely used in China as a treatment for cough, bleeding, and gastrointestinal disorders. In recent years, the anticancer activity of GA has been demonstrated in a variety of cancers, affecting multiple cellular pathways associated with cancer onset, development and progression. AIM OF THE STUDY: To investigate the role and potential mechanism of GA on gastric precancerous lesions (GPL), the key turning point of gastritis to gastric cancer, with the aim of delaying, blocking or reversing the dynamic overall process of "inflammation-cancer transformation" and thus blocking GPL to prevent the development of gastric cancer. MATERIALS AND METHODS: In this study, we established N-Nitroso-N-methylurea (MNU)-induced GPL mice model and induced precancerous lesions of gastric cancer cells (MC), i.e. epithelial mesenchymal transition (EMT), in human gastric mucosal epithelial cells (GES-1) with N-methyl-N'-nitro-N-nitrosoguanidine (MNNG). We used conventional pathology, immunohistochemistry, RNA sequencing, Western blot and other techniques to study the therapeutic effect of GA on GPL and its possiblemechanism in vitro and in vivo. RESULTS: The results showed that compared with normal GES-1 cells, MC cells had the characteristics of malignant cells such as abnormal proliferation, invasion and metastasis, accompanied by decreased expression of EMT-related protein E-cadherin and increased expression of N-cadherin and Vimentin. GA can inhibit the malignant behavior of MC cell proliferation and induce its G0/G1 phase arrest, which is achieved by downregulating the Wnt/ß-catenin signaling pathway and thereby inhibiting the EMT process. However, when we incubated with the Wnt pathway activator (Wnt agonist 1), the effect of GA was reversed. Furthermore, analysis of human gastric specimens showed that activation of the Wnt/ß-catenin pathway was significantly associated with GPL pathological changes. Meanwhile, GA reversed MNU-induced intestinal metaplasia and partial dysplasia in GPL mice. CONCLUSION: Taken together, these results indicate that GA prevents the occurrence and development of GPL by inhibiting the Wnt/ß-catenin signaling pathway and then inhibiting the EMT process, which may become potential candidates for the treatment of GPL.


Assuntos
Lesões Pré-Cancerosas , Neoplasias Gástricas , Humanos , Camundongos , Animais , Via de Sinalização Wnt , Transição Epitelial-Mesenquimal , Neoplasias Gástricas/genética , Ácido Gálico/farmacologia , Ácido Gálico/uso terapêutico , Tosse , Movimento Celular , beta Catenina/metabolismo , Proliferação de Células , Lesões Pré-Cancerosas/induzido quimicamente , Lesões Pré-Cancerosas/tratamento farmacológico , Metilnitronitrosoguanidina , Caderinas/metabolismo , Linhagem Celular Tumoral
13.
Phytomedicine ; 108: 154524, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36375238

RESUMO

BACKGROUND: Cancer stem cells (CSCs) are strongly associated with high tumourigenicity, chemotherapy or radiotherapy resistance, and metastasis and recurrence, particularly in colorectal cancer (CRC). Therefore, targeting CSCs may be a promising approach. Recently, discovery and research on phytochemicals that effectively target colorectal CSCs have been gaining popularity because of their broad safety profile and multi-target and multi-pathway modes of action. PURPOSE: This review aimed to elucidate and summarise the effects and mechanisms of phytochemicals with potential anti-CSC agents that could contribute to the better management of CRC. METHODS: We reviewed PubMed, EMBASE, Web of Science, Ovid, ScienceDirect and China National Knowledge Infrastructure databases from the original publication date to March 2022 to review the mechanisms by which phytochemicals inhibit CRC progression by targeting CSCs and their key signalling pathways. Phytochemicals were classified and summarised based on the mechanisms of action. RESULTS: We observed that phytochemicals could affect the biological properties of colorectal CSCs. Phytochemicals significantly inhibit self-renewal, migration, invasion, colony formation, and chemoresistance and induce apoptosis and differentiation of CSCs by regulating the Wnt/ß-catenin pathway (e.g., diallyl trisulfide and genistein), the phosphatidylinositol-3-kinase/protein kinase B/mammalian target of rapamycin pathway (e.g., caffeic acid and piperlongumine), the neurogenic locus notch homolog protein pathway (e.g., honokiol, quercetin, and α-mangostin), the Janus kinase-signal transducer and activator of transcription pathway (e.g., curcumin, morin, and ursolic acid), and other key signalling pathways. It is worth noting that several phytochemicals, such as resveratrol, silibinin, evodiamine, and thymoquinone, highlight multi-target and multi-pathway effects in restraining the malignant biological behaviour of CSCs. CONCLUSIONS: This review demonstrates the potential of targeted therapies for colorectal CSCs using phytochemicals. Phytochemicals could serve as novel therapeutic agents for CRC and aid in drug development.


Assuntos
Neoplasias Colorretais , Células-Tronco Neoplásicas , Humanos , Compostos Fitoquímicos/uso terapêutico , Via de Sinalização Wnt , Apoptose , Receptores Notch/metabolismo , Neoplasias Colorretais/patologia
14.
Dis Markers ; 2022: 4492608, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36168326

RESUMO

Objective: Colon adenocarcinoma (COAD) is one of the most prevalent cancers worldwide. However, the pyroptosis-related lncRNAs of COAD have not been deeply examined and validated. Here, we constructed and validated a risk model on pyroptosis-related lncRNAs in COAD. Methods: The RNA sequencing transcriptome and clinical data of COAD patients were downloaded from The Cancer Genome Atlas (TCGA) database. Differentially expressed pyroptosis-related mRNAs and mRNA-lncRNA coexpression network were identified. After univariate and multifactorial cox analyses of prognosis-related lncRNAs, a risk model was constructed. Next, we analyzed the differences in immune infiltration, immune checkpoint blockade-, immune checkpoint-, and N6-methyladenosine-related gene expressions between the high- and low-risk groups. RT-qPCR was used to validate the expression of lncRNAs. Result: A risk model was constructed based on 9 pyroptosis-related lncRNAs and separated COAD patients into the high- and low-risk groups. Immune infiltration analysis and immune checkpoint blockade-, immune checkpoint-, and N6-methyladenosine-related genes showed significant differences between the two subgroups. RT-qPCR showed that the 9 pyroptosis-related lncRNAs could be used as prognostic indicators. Conclusion: A novel risk model based on pyroptosis-related lncRNAs was constructed and demonstrated that these lncRNAs might be used as independent prognostic biomarkers. This will also assist shed light on the COAD prognosis and therapy.


Assuntos
Adenocarcinoma , Neoplasias do Colo , RNA Longo não Codificante , Adenocarcinoma/genética , Adenocarcinoma/patologia , Biomarcadores Tumorais/genética , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Redes Reguladoras de Genes , Humanos , Inibidores de Checkpoint Imunológico , Prognóstico , Piroptose/genética , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
15.
Pharmacol Res ; 184: 106440, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36108874

RESUMO

Gastric carcinoma (GC) is a complex multifactorial disease occurring as sequential events commonly referred to as the Correa's cascade, a stepwise progression from non-active or chronic active gastritis, to gastric precancerous lesions, and finally, adenocarcinoma. Therefore, the identification of novel agents with multi-step actions on the Correa's cascade and those functioning as multiple phenotypic regulators are the future direction for drug discovery. Recently, berberine (BBR) has gained traction owing to its pharmacological properties, including anti-inflammatory, anti-cancer, anti-ulcer, antibacterial, and immunopotentiation activities. In this article, we investigated and summarized the multi-step actions of BBR on Correa's cascade and its underlying regulatory mechanism in gastric carcinogenesis for the first time, along with a discussion on the strength of BBR to prevent and treat GC. BBR was found to suppress H. pylori infection, control mucosal inflammation, and promote ulcer healing. In the gastric precancerous lesion phase, BBR could reverse mucosal atrophy and prevent lesions in intestinal metaplasia and dysplasia by regulating inflammatory cytokines, promoting cell apoptosis, regulating macrophage polarization, and regulating autophagy. Additionally, the therapeutic action of BBR on GC was partly realized through the inhibition of cell proliferation, migration, and angiogenesis; induction of apoptosis and autophagy, and enhancement of chemotherapeutic drug sensitivity. BBR exerted multi-step actions on the Correa's cascade, thereby halting and even reversing gastric carcinogenesis in some cases. Thus, BBR could be used to prevent and treat GC. In conclusion, the therapeutic strategy underlying BBR's multi-step action in the trilogy of Correa's cascade may include "prevention of gastric mucosal inflammation (Phase 1); reversal of gastric precancerous lesions (Phase 2), and rescue of GC (Phase 3)". The NF-κB, PI3K/Akt, and MAPK signaling pathways may be the key signaling transduction pathways underlying the treatment of gastric carcinogenesis using BBR. The advantage of BBR over conventional drugs is its multifaceted and long-term effects. This review is expected to provide preclinical evidence for using BBR to prevent gastric carcinogenesis and treat gastric cancer.


Assuntos
Berberina , Gastrite Atrófica , Gastrite , Infecções por Helicobacter , Lesões Pré-Cancerosas , Neoplasias Gástricas , Antibacterianos/uso terapêutico , Berberina/farmacologia , Berberina/uso terapêutico , Carcinogênese , Citocinas/uso terapêutico , Gastrite Atrófica/patologia , Infecções por Helicobacter/complicações , Infecções por Helicobacter/tratamento farmacológico , Infecções por Helicobacter/microbiologia , Humanos , Inflamação , NF-kappa B , Fosfatidilinositol 3-Quinases , Lesões Pré-Cancerosas/patologia , Proteínas Proto-Oncogênicas c-akt , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/patologia , Neoplasias Gástricas/prevenção & controle
16.
Int J Nanomedicine ; 17: 4163-4193, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36134202

RESUMO

Cancer stem cells (CSCs) lead to the occurrence and progression of cancer due to their strong tumorigenic, self-renewal, and multidirectional differentiation abilities. Existing cancer treatment methods cannot effectively kill or inhibit CSCs but instead enrich them and produce stronger proliferation, invasion, and metastasis capabilities, resulting in cancer recurrence and treatment resistance, which has become a difficult problem in clinical treatment. Therefore, targeting CSCs may be the most promising approach for comprehensive cancer therapy in the future. A variety of natural products (NP) have significant antitumor effects and have been identified to target and inhibit CSCs. However, pharmacokinetic defects and off-target effects have greatly hindered their clinical translation. NP-based nanoformulations (NPNs) have tremendous potential to overcome the disadvantages of NP against CSCs through site-specific delivery and by improving their pharmacokinetic parameters. In this review, we summarize the recent progress of NPNs targeting CSCs in cancer therapy, looking forward to transforming preclinical research results into clinical applications and bringing new prospects for cancer treatment.


Assuntos
Produtos Biológicos , Neoplasias , Produtos Biológicos/farmacologia , Produtos Biológicos/uso terapêutico , Diferenciação Celular , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Células-Tronco Neoplásicas/patologia
17.
Front Pharmacol ; 13: 797805, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35846998

RESUMO

Background: Blocking and even reversing gastric precancerous lesions (GPL) is a key measure to lower the incidence of gastric cancer. Atractylenolide III (AT-III) is a mainly active component of the Atractylodes rhizome and has been widely used in tumor treatment. However, the effects of AT-III on GPL and its mechanisms have not been reported. Methods: H & E staining and AB-PAS staining were employed to evaluate the histopathology in the gastric mucosa. In parallel, CD34 immunostaining was performed for angiogenesis assessment, and transmission electron microscope for microvessel ultrastructural observation. Investigation for the possible mechanism in vivo and in vitro was conducted using immunohistochemistry, RT-qPCR and western blotting. Results: In most GPL specimens, AT-III treatment reduced microvascular abnormalities and attenuated early angiogenesis, with the regression of most intestinal metaplasia and partial dysplasia. Meanwhile, the expression of VEGF-A and HIF-1α was enhanced in GPL samples of model rats, and their expressions were decreased in AT-III-treated GPL rats. Moreover, DLL4 mRNA and protein expression were higher in GPL rats than in control rats. DLL4 protein expression was significantly enhanced in human GPL tissues. In addition, AT-III treatment could diminish DLL4 mRNA level and protein expression in the MNNG-induced GPL rats. In vitro study showed that in AGS and HGC-27 cells, DLL4 mRNA level and protein expression were significantly decreased after AT-III treatment. However, AT-III had no significant regulatory effect on Notch1 and Notch4. Conclusion: AT-III treatment is beneficial in lessening gastric precancerous lesions and attenuating angiogenesis in rats, and that may be contributed by the decrease of angiogenesis-associated HIF-1α and VEGF-A, and downregulation of DLL4.

18.
Front Pharmacol ; 13: 906301, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35721116

RESUMO

In many studies, the extensive and significant anticancer activity of chelerythrine (CHE) was identified, which is the primary natural active compound in four traditional botanical drugs and can be applied as a promising treatment in various solid tumors. So this review aimed to summarize the anticancer capacities and the antitumor mechanism of CHE. The literature searches revolving around CHE have been carried out on PubMed, Web of Science, ScienceDirect, and MEDLINE databases. Increasing evidence indicates that CHE, as a benzophenanthridine alkaloid, exhibits its excellent anticancer activity as CHE can intervene in tumor progression and inhibit tumor growth in multiple ways, such as induction of cancer cell apoptosis, cell cycle arrest, prevention of tumor invasion and metastasis, autophagy-mediated cell death, bind selectively to telomeric G-quadruplex and strongly inhibit the telomerase activity through G-quadruplex stabilization, reactive oxygen species (ROS), mitogen-activated protein kinase (MAPK), and PKC. The role of CHE against diverse types of cancers has been investigated in many studies and has been identified as the main antitumor drug candidate in drug discovery programs. The current complex data suggest the potential value in clinical application and the future direction of CHE as a therapeutic drug in cancer. Furthermore, the limitations and the present problems are also highlighted in this review. Despite the unclearly delineated molecular targets of CHE, extensive research in this area provided continuously fresh data exploitable in the clinic while addressing the present requirement for further studies such as toxicological studies, combination medication, and the development of novel chemical methods or biomaterials to extend the effects of CHE or the development of its derivatives and analogs, contributing to the effective transformation of this underestimated anticancer drug into clinical practice. We believe that this review can provide support for the clinical application of a new anticancer drug in the future.

19.
Biomed Pharmacother ; 145: 112086, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34799220

RESUMO

Ginsenoside Rg3 (GRg3) is a ginsenoside extracted from Panax ginseng. GRg3 displays multiple pharmacological properties, such as antitumor, anti-inflammatory, antioxidative and antifibrotic properties. However, whether GRg3 inhibits angiogenesis in gastric precancerous lesions (GPLs) and the possible mechanisms remain unknown. GRg3 attenuated gastric intestinal metaplasia and gastric dysplasia, the hallmark of GPL pathology, in rats with MNNG-ammonia compound induced GPLs. Increased CD34+ microvessel density and VEGF expression, which indicate the presence of angiogenesis, were evident in the rats with GPLs. GRg3 administration reduced VEGF protein expression and CD34+ microvessel density. In addition, GRg3 was capable of attenuating microvascular abnormalities. Data analysis revealed that enhanced protein expression of GLUT1, GLUT3 and GLUT4 were present in both human and animal GPL specimens. The administration of GRg3 caused significant decreases in the mRNA and protein expression levels of GLUT1 and GLUT4 in the rats with GPLs. However, the GRg3-treated rats with GPLs did not demonstrate regulatory effects on GLUT3, GLUT6, GLUT10, and GLUT12. Consistent with in vitro results, GRg3 administration significantly reduced the protein expression levels of GLUT1 and GLUT4 in both AGS and HGC-27 human gastric cancer cells in vitro. In conclusion, GRg3 can attenuate angiogenesis and temper microvascular abnormalities in rats with GPLs, which may be associated with its inhibition on the aberrant activation of GLUT1 and GLUT4.


Assuntos
Ginsenosídeos/farmacologia , Neovascularização Patológica/tratamento farmacológico , Lesões Pré-Cancerosas/tratamento farmacológico , Neoplasias Gástricas/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Transportador de Glucose Tipo 1/genética , Transportador de Glucose Tipo 4/genética , Humanos , Masculino , Neovascularização Patológica/genética , Lesões Pré-Cancerosas/genética , Ratos , Ratos Sprague-Dawley , Estudos Retrospectivos , Neoplasias Gástricas/genética
20.
Front Oncol ; 11: 775418, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34869024

RESUMO

BACKGROUND: Cancer stem cells (CSCs) are able to survive after cancer therapies, resulting in tumor progression and recurrence, as is seen in colorectal cancer. Therapies targeting CSCs are regarded as novel and promising strategies for efficiently eradicating tumors. Berberine, an isoquinoline alkaloid extracted from the Chinese herbal medicine Coptis chinensis, was found to have antitumor activities against colorectal cancer, without knowing whether it exerts inhibitory effects on colorectal CSCs and the potential mechanisms. METHODS: In this study, we examined the inhibitory roles of Berberine on CSCs derived from HCT116 and HT29 by culturing in serum-free medium. We also examined the effects of Berberine on m6A methylation via regulating fat mass and obesity-associated protein (FTO), by downregulating ß-catenin. RESULTS: We examined the effects of Berberine on the tumorigenicity, growth, and stemness of colorectal cancer stem-like cells. The regulatory effect of Berberine on N6-methyladenosine (m6A), an abundant mRNA modification, was also examined. Berberine treatment decreased cell proliferation by decreasing cyclin D1 and increasing p27 and p21 and subsequently induced cell cycle arrest at the G1/G0 phase. Berberine treatment also decreased colony formation and induced apoptosis. Berberine treatment transcriptionally increased FTO and thus decreased m6A methylation, which was reversed by both FTO knockdown and the addition of the FTO inhibitor FB23-2. Berberine induced FTO-related decreases in stemness in HCT116 and HT29 CSCs. Berberine treatment also increased chemosensitivity in CSCs and promoted chemotherapy agent-induced apoptosis. Moreover, we also found that Berberine treatment increased FTO by decreasing ß-catenin, which is a negative regulator of FTO. CONCLUSIONS: Our observation that Berberine effectively decreased m6A methylation by decreasing ß-catenin and subsequently increased FTO suggests a role of Berberine in modulating stemness and malignant behaviors in colorectal CSCs.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...