Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Transl Med ; 22(1): 249, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38454407

ABSTRACT

BACKGROUND: Bioactive lipids involved in the progression of various diseases. Nevertheless, there is still a lack of biomarkers and relative regulatory targets. The lipidomic analysis of the samples from platinum-resistant in gastric cancer patients is expected to help us further improve our understanding of it. METHODS: We employed LC-MS based untargeted lipidomic analysis to search for potential candidate biomarkers for platinum resistance in GC patients. Partial least squares discriminant analysis (PLS-DA) and variable importance in projection (VIP) analysis were used to identify differential lipids. The possible molecular mechanisms and targets were obtained by metabolite set enrichment analysis and potential gene network screened. Finally, verified them by immunohistochemical of a tissue microarray. RESULTS: There were 71 differential lipid metabolites identified in GC samples between the chemotherapy-sensitivity group and the chemotherapy resistance group. According to Foldchange (FC) value, VIP value, P values (FC > 2, VIP > 1.5, p < 0.05), a total of 15 potential biomarkers were obtained, including MGDG(43:11)-H, Cer(d18:1/24:0) + HCOO, PI(18:0/18:1)-H, PE(16:1/18:1)-H, PE(36:2) + H, PE(34:2p)-H, Cer(d18:1 + hO/24:0) + HCOO, Cer(d18:1/23:0) + HCOO, PC(34:2e) + H, SM(d34:0) + H, LPC(18:2) + HCOO, PI(18:1/22:5)-H, PG(18:1/18:1)-H, Cer(d18:1/24:0) + H and PC(35:2) + H. Furthermore, we obtained five potential key targets (PLA2G4A, PLA2G3, DGKA, ACHE, and CHKA), and a metabolite-reaction-enzyme-gene interaction network was built to reveal the biological process of how they could disorder the endogenous lipid profile of platinum resistance in GC patients through the glycerophospholipid metabolism pathway. Finally, we further identified PLA2G4A and ACHE as core targets of the process by correlation analysis and tissue microarray immunohistochemical verification. CONCLUSION: PLA2G4A and ACHE regulated endogenous lipid profile in the platinum resistance in GC patients through the glycerophospholipid metabolism pathway. The screening of lipid biomarkers will facilitate earlier precision medicine interventions for chemotherapy-resistant gastric cancer. The development of therapies targeting PLA2G4A and ACHE could enhance platinum chemotherapy effectiveness.


Subject(s)
Stomach Neoplasms , Humans , Biomarkers , Discriminant Analysis , Glycerophospholipids , Group III Phospholipases A2 , Group IV Phospholipases A2 , Lipid Metabolism/genetics , Lipids , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics
2.
Phytomedicine ; 128: 155360, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38547624

ABSTRACT

BACKGROUND: Autophagy could sense metabolic conditions and safeguard cells against nutrient deprivation, ultimately supporting the survival of cancer cells. Nobiletin (NOB) is a kind of bioactive component of the traditional Chinese medicine Citri Reticulatae Pericarpium and has been proven to induce GC cell death by reducing de novo fatty acid synthesis in our previous study. Nevertheless, the precise mechanisms by which NOB induces cell death in GC cells still need further elucidation. OBJECTIVES: To examine the mechanism by which NOB inhibits gastric cancer progression through the regulation of autophagy under the condition of lipid metabolism inhibition. METHODS/ STUDY DESIGN: Proliferation was detected by the CCK-8 assay. RNA sequencing (RNA-seq) was used to examine signaling pathway changes. Electron microscopy and mRFP-GFP-LC3 lentiviral transfection were performed to observe autophagy in vitro. Western blot, plasmid transfection, immunofluorescence staining, and CUT & Tag-qPCR techniques were utilized to explore the mechanisms by which NOB affects GC cells. Molecular docking and molecular dynamics simulations were conducted to predict the binding mode of NOB and SREBP1. CETSA was adopted to verify the predicted of binding model. A patient-derived xenograft (PDX) model was employed to verify the therapeutic efficacy of NOB in vivo. RESULTS: We conducted functional studies and discovered that NOB inhibited the protective effect of autophagy via the PI3K/Akt/mTOR axis in GC cells. Based on previous research, we found that the overexpression of ACLY abrogated the NOB-induced autophagy-dependent cell death. In silico analysis predicted the formation of a stable complex between NOB and SREBP1. In vitro assays confirmed that NOB treatment increased the thermal stability of SREBP1 at the same temperature conditions. Moreover, CUT&TAG-qPCR analysis revealed that NOB could inhibit SREBP1 binding to the ACLY promoter. In the PDX model, NOB suppressed tumor growth, causing SREBP1 nuclear translocation inhibition, PI3K/Akt/mTOR inactivation, and autophagy-dependent cell death. CONCLUSION: NOB demonstrated the ability to directly bind to SREBP1, inhibiting its nuclear translocation and binding to the ACLY promoter, thereby inducing autophagy-dependent cell death via PI3K/Akt/mTOR pathway.


Subject(s)
Autophagy , Flavones , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , Sterol Regulatory Element Binding Protein 1 , Stomach Neoplasms , TOR Serine-Threonine Kinases , Humans , Stomach Neoplasms/drug therapy , TOR Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Flavones/pharmacology , Sterol Regulatory Element Binding Protein 1/metabolism , Animals , Autophagy/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Cell Line, Tumor , Molecular Docking Simulation , Mice , Mice, Nude , Mice, Inbred BALB C
3.
J Adv Res ; 2024 Jan 29.
Article in English | MEDLINE | ID: mdl-38295877

ABSTRACT

INTRODUCTION: Dehydrocostus lactone (Dehy), a natural sesquiterpene lactone from Saussurea lappa Clarke, displays remarkable efficacy in treating cancer and gastrointestinal disorders. However, its anti-gastric cancer (GC) effect remains poorly understood. OBJECTIVES: Our study aimed to elucidate the anti-GC effect of Dehy and its putative mechanism. METHODS: The anti-GC effect was assessed with MTT, colony formation, wound healing and transwell invasion assays. Cell apoptosis rate was detected by Annexin V-FITC/PI binding assay. Network pharmacology analysis and XF substrate oxidation stress test explored the underlying mechanism and altered metabolic phenotype. Lipogenic enzyme expressions and neutral lipid pool were measured to evaluate cellular lipid synthesis and storage. Biolayer interferometry and molecular docking investigated the direct target of Dehy. Autophagosomes were observed by transmission electron microscopy and MDC staining, while the autophagic flux was detected by mRFP-GFP-LC3 transfection. The clinical significance of ACLY was confirmed by tissue microarrays. Patient-derived xenograft (PDX) models were adopted to detect the clinical therapeutic potential of Dehy. RESULTS: Dehy prominently suppressed GC progression both in vitro and in vivo. Mechanistically, Dehy down-regulated the lipogenic enzyme ACLY, thereby reducing fatty acid synthesis and lipid reservation. Moreover, IKKß was identified as the direct target of Dehy. Dehy inhibited the phosphorylation of IKKß, promoting the ubiquitination and degradation of ACLY, thereby resulting in lipid depletion. Subsequently, GC cells initiated autophagy to replenish the missing lipids, whereas Dehy impeded this cytoprotective mechanism by down-regulating LAMP1 and LAMP2 expressions, which disrupted lysosomal membrane functions, ultimately leading to apoptosis. Additionally, Dehy exhibited potential in GC clinical therapy as it enhanced the efficacy of 5-Fluorouracil in PDX models. CONCLUSIONS: Our work identified Dehy as a desirable agent for blunting abnormal lipid metabolism and highlighted its inhibitory effect on protective autophagy, suggesting the future development of Dehy as a novel therapeutic drug for GC.

4.
BMC Complement Med Ther ; 23(1): 295, 2023 Aug 22.
Article in English | MEDLINE | ID: mdl-37608281

ABSTRACT

BACKGROUND: Saikosaponin A (SSA) shows a series of pharmacological activities, such as anti-inflammatory, antioxidant and antitumor. However, there is a lack of comprehensive research or sufficient evidence regarding the efficacy of SSA in treating gastric cancer (GC), and the specific mechanisms by which it inhibits GC growth and progression are still not fully understood. METHODS: MTT and clonogenic assays were employed to detect the effect of SSA on the proliferation of GC cells. Bioinformatics predicted the SSA targets in the treatment of GC. The core genes and the underlying mechanism of SSA in anti-GC were obtained by analyzing the intersecting targets; molecular docking and Western blot were used to check the reliability of core genes. Flow cytometry was used to analyze apoptosis and cell cycle in GC cells treated with varying concentrations of SSA. Western blot was employed to detect the expression levels of related proteins. RESULTS: SSA significantly blocked GC cells in the S phase of the cell cycle and induced apoptosis to suppress the proliferation of GC cells. Network pharmacology revealed that the underlying mechanisms through which SSA acts against GC involve the modulation of several signaling pathways, including the PI3K-Akt, MAPK, RAS, and T-cell signaling pathways. Molecular docking showed pivotal target genes with a high affinity to SSA, including STAT3, MYC, TNF, STAT5B, Caspase-3 and SRC. Furthermore, western blot results revealed that SSA significantly increased the protein levels of Bax and Cleaved Caspase-3, whereas decreased the expression levels of p-JAK, p-STAT3, MYC, Bcl-2, p-PI3K, p-AKT and p-mTOR, confirming that the reliability of hub targets and SSA could promote GC cell apoptosis by suppressing PI3K/AKT/mTOR pathway. CONCLUSIONS: The results suggest that SSA has the ability to trigger apoptosis in GC cells by blocking the PI3K/AKT/mTOR pathway. These findings highlight the potential of SSA as a promising natural therapeutic agent for the treatment of GC.


Subject(s)
Stomach Neoplasms , Humans , Stomach Neoplasms/drug therapy , Caspase 3 , Molecular Docking Simulation , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Reproducibility of Results
5.
Phytomedicine ; 116: 154902, 2023 Jul 25.
Article in English | MEDLINE | ID: mdl-37270969

ABSTRACT

BACKGROUND: Gastric cancer (GC) is a common malignant tumor with limited treatment options. The natural flavonoid nobiletin (NOB) is a beneficial antioxidant that possesses anticancer activity. However, the mechanisms by which NOB inhibits GC progression remain unclear. METHODS: A CCK-8 assay was performed to determine cytotoxicity. Cell cycle and apoptosis analyses were performed by flow cytometry. RNA-seq was performed to detect differential gene expression after NOB treatment. RT‒qPCR, Western blot and immunofluorescence staining were used to examine the underlying mechanisms of NOB in GC. Xenograft tumor models were constructed to verify the effect of NOB and its specific biological mechanism in GC. RESULTS: NOB inhibited cell proliferation, caused cell cycle arrest and induced apoptosis in GC cells. KEGG classification identified that the inhibitory effect of NOB on GC cells mainly involved the lipid metabolism pathway. We further showed that NOB reduced de novo fatty acid (FA) synthesis, as evidenced by the decreased levels of neutral lipids and the expression levels of ACLY, ACACA and FASN, and ACLY abrogated the effect of NOB on lipid deposits in GC cells. In addition, we also found that NOB triggered endoplasmic reticulum (ER) stress by activating the IRE-1α/GRP78/CHOP axis, but overexpression of ACLY reversed ER stress. Mechanistically, inhibiting ACLY expression with NOB significantly reduced neutral lipid accumulation, thereby inducing apoptosis by activating IRE-1α-mediated ER stress and inhibiting GC cell progression. Finally, in vivo results also demonstrated that NOB inhibited tumor growth by decreasing de novo FA synthesis. CONCLUSION: NOB could inhibit the expression of ACLY to activate IRE-1α-induced ER stress, which ultimately led to GC cell apoptosis. Our results provide novel insight into the use of de novo FA synthesis for GC treatment and are the first to reveal that NOB inhibits GC progression by ACLY-dependent ER stress.


Subject(s)
Stomach Neoplasms , Animals , Humans , Stomach Neoplasms/genetics , Cell Line, Tumor , Disease Models, Animal , Endoplasmic Reticulum Stress , Lipids , Apoptosis
6.
J Ethnopharmacol ; 311: 116450, 2023 Jul 15.
Article in English | MEDLINE | ID: mdl-37023839

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Developing complementary and effective drugs with less toxicity is urgent for gastric cancer (GC) therapy. Jianpi Yangzheng Decoction (JPYZ) is a curative medical plants formula against GC in clinic while its molecular mechanism remains to be further elucidated. AIM OF THE STUDY: To evaluate the in vitro and in vivo anticancer efficacy of JPYZ against GC and its potential mechanisms. MATERIALS AND METHODS: The effect of JPYZ on regulating the candidate targets were screened and examined by RNA-Seq, qRT-PCR, luciferase reporter assay, and immunoblotting. Rescue experiment was conducted to authenticate the regulation of JPYZ on the target gene. Molecular interaction, intracellular localization and function of target genes were elucidated via Co-IP and cytoplasmic-nuclear fractionation. The impact of JPYZ on the abundance of target gene in clinical specimens of GC patients was evaluated by IHC. RESULTS: JPYZ treatment suppressed the proliferation and metastasis of GC cells. RNA seq revealed JPYZ significantly downregulated miR-448. A reporter plasmid containing CLDN18 3'-UTR WT exhibited significant decrease in luciferase activity when co-transfected with miR-448 mimic in GC cells. CLDN18.2 deficiency promoted the proliferation and metastasis of GC cells in vitro, as well as intensified the growth of GC xenograft in mice. JPYZ reduced the proliferation and metastasis of GC cells with CLDN18.2 abrogation. Mechanically, suppressed activities of transcriptional coactivator YAP/TAZ and its downstream targets were observed in GC cells with CLDN18.2 overexpression and those under JPYZ treatment, leading to cytoplasmic retention of phosphorylated YAP at site Ser-127. High abundance of CLDN18.2 was detected in more GC patients who received chemotherapy combined with JPYZ. CONCLUSION: JPYZ has an inhibitory effect on GC growth and metastasis partly by elevating CLDN18.2 abundance in GC cells, indicating more patients may benefit from combination therapy of JPYZ and the upcoming CLDN18.2 target agents.


Subject(s)
MicroRNAs , Stomach Neoplasms , Humans , Animals , Mice , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Signal Transduction , Transcription Factors/genetics , Cell Line, Tumor , MicroRNAs/genetics , Cell Proliferation , Gene Expression Regulation, Neoplastic , Claudins/genetics , Claudins/metabolism
7.
Phytomedicine ; 103: 154229, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35691076

ABSTRACT

BACKGROUND: Modified Jianpi Yangzheng decoction (mJPYZ), as an empirical decoction of Traditional Chinese medicine has been shown significantly to prolong the survival of patients with advanced stage gastric cancer. Pyruvate kinase M2 (PKM2), has attracted attention for its important role on cellular aerobic glycolysis, however, few studies focus on PKM2 non-metabolic roles in tumor progression. PURPOSE: Our study aimed to investigate the potential role of gastric cancer exosomes containing PKM2 in regulating tumor-associated macrophages (TAM) and the mechanism of mJPYZ against gastric cancer. METHODS: Colony Formation Assay, flow cytometry and TUNEL staining were employed to estimate the effect of mJPYZ on gastric cancer in tumor-bearing mice and cells. Western blot analyzed apoptosis-related protein expression changes. Network pharmacology and bioinformatics predicted potential exosomes modulation of mJPYZ in gastric cancer. Exosomes were isolated and co-cultured with TAM. Diff-Quik Staining observed the TAM morphological changes when incubating with gastric cancer cells exosomes. Flow cytometry and immunofluorescence were performed to demonstrate whether exosomes PKM2 involved in TAM polarization. RESULTS: mJPYZ induced apoptosis of gastric cancer cells by targeting PKM2 and downregulating PI3K/Akt/mTOR axis in vivo and in vitro. Network pharmacology showed potential exosomes modulation of mJPYZ in gastric cancer. We extracted exosomes and found mJPYZ decreased the abundance of serum exosomes PKM2 in patients with advanced gastric cancer and xenograft tumor model. Additionally, we firstly detected and confirmed that PKM2 is a package protein of exosomes extracted from gastric cancer cells, and mJPYZ could diminish the content of exosomal PKM2 in gastric cancer cells. Importantly, mJPYZ reduced the delivery of exosomal PKM2 from tumor cells to macrophages, and alleviated exosomal PKM2-induced differentiation of M2-TAM in tumor microenvironment, eventually inhibited gastric cancer progression. CONCLUSION: Gastric cancer exosomes containing PKM2 could lead to M2 macrophages differentiation, thereby promoting gastric cancer progression. Our findings provide a rationale for potential application of mJPYZ in the treatment of gastric cancer via PKM2.


Subject(s)
Drugs, Chinese Herbal , Exosomes , Pyruvate Kinase , Stomach Neoplasms , Animals , Carrier Proteins/metabolism , Cell Line, Tumor , Drugs, Chinese Herbal/pharmacology , Exosomes/drug effects , Exosomes/metabolism , Exosomes/pathology , Humans , Membrane Proteins/metabolism , Mice , MicroRNAs/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Pyruvate Kinase/metabolism , Stomach Neoplasms/drug therapy , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology , Thyroid Hormones/metabolism , Tumor Microenvironment , Thyroid Hormone-Binding Proteins
8.
Front Oncol ; 11: 767116, 2021.
Article in English | MEDLINE | ID: mdl-34926270

ABSTRACT

Gastric cancer is the third leading cause of cancer death worldwide. Traditional Chinese medicine (TCM) is increasingly extensively applied as a complementary therapy for gastric cancer (GC) in China, which shows unique advantages in preventing gastric cancer metastasis. Previous study indicates modified Jian-pi-yang-zheng (mJPYZ) decoction inhibit the progression of gastric cancer by regulating tumor-associated macrophages (TAM). However, it is unclear whether mJPYZ can affect metabolic reprogramming of gastric cancer cells. Here, we showed that mJPYZ effectively attenuated GC cells proliferation, migration and invasion. Meantime, mJPYZ reduced the aerobic glycolysis level of GC cells in vivo and in vitro by regulating the expression and nuclear translocation of PKM2. Overexpression of PKM2 that could reverse the inhibitory effect of mJPYZ, migration and epithelial to mesenchymal transition (EMT). Our results showed PKM2/HIF-1α signaling was the key metabolic regulator of mJPYZ in GC cells. In summary, our present study suggested that abnormal PKM2 is required for maintaining the malignant phenotype of GC cells. The TCM decoction mJPYZ inhibited GC cells growth and EMT by reducing of glycolysis in PKM2 dependent manner. This evidence expanded our understanding of the anti-tumor mechanism of mJPYZ and further indicated mJPYZ a potential anti-tumor agent for GC patients. CHEMICAL COMPOUNDS STUDIED IN THIS ARTICLE: Rutin (PubChem CID: 5280805); Lobetyolin (PubChem CID: 53486204); Calycosin-7-glucoside (PubChem CID: 71571502); Formononetin (PubChem CID: 5280378); Calycosin (PubChem CID: 5280448); Ononin (PubChem CID: 442813); P-Coumaric Acid (PubChem CID: 637542).

9.
Inorg Chem ; 60(21): 16016-16028, 2021 Nov 01.
Article in English | MEDLINE | ID: mdl-34633179

ABSTRACT

A comprehensive experimental and theoretical study of both thermal-induced spin transition (TIST) as a function of pressure and pressure-induced spin transition (PIST) at room temperature for the two-dimensional Hofmann-like SCO polymer [Fe(Fpz)2Pt(CN)4] is reported. The TIST studies at different fixed pressures have been carried out by magnetic susceptibility measurements, while PIST studies have been performed by means of powder X-ray diffraction, Raman, and visible spectroscopies. A combination of the theory of elastic interactions and numerical Monte Carlo simulations has been used for the analysis of the cooperative interactions in TIST and PIST studies. A complete (T, P) phase diagram for the compound [Fe(Fpz)2Pt(CN)4] has been constructed. The critical temperature of the spin transition follows a lineal dependence with pressure, meanwhile the hysteresis width shows a nonmonotonic behavior contrary to theoretical predictions. The analysis shows the exceptional role of the total entropy and phonon contribution in setting the temperature of the spin transition and the width of the hysteresis. The anomalous behavior of the thermal hysteresis width under pressure in [Fe(Fpz)2Pt(CN)4] is a direct consequence of a local distortion of the octahedral geometry of the Fe(II) centers for pressures higher than 0.4 GPa. Interestingly, there is not a coexistence of the high- and low-spin (HS and LS, respectively) phases in TIST experiments, while in PIST experiments, the coexistence of the HS and LS phases in the metastable region of the phase transition induced by pressure is observed for a first time in a first-order gradual spin transition with hysteresis.

10.
Curr Drug Targets ; 22(11): 1222-1231, 2021.
Article in English | MEDLINE | ID: mdl-33845736

ABSTRACT

Gut microbiota is a complex microecosystem, which is called the second genome of the human body. Herbal medicine can balance tumor-suppressing bacteria and tumor-promoting bacteria and exert its anti-cancer effect by regulating gut microbiota. Traditional Chinese medicine (TCM) has a history of thousands of years in prevention and treatment of diseases in China. In recent decades, TCM has been shown to have an obvious advantage in prolonging the survival time and improving the living quality of patients with cancer. Notably, gut microbiota has become a new pathway to understanding TCM. In this review, we will focus on gut microbiota and tumor progression, especially the diversity, functionality and metabolites of gut microbiota affected by TCM in various cancer. We will also discuss the potential mechanism of gut microbiota for exploring TCM in anti-cancer effect. This article aims to comprehensively review the anti-cancer research of TCM by regulating gut microbiota, and address future perspectives and challenges of gut microbiota in TCM intervention for cancer.


Subject(s)
Drugs, Chinese Herbal , Gastrointestinal Microbiome , Neoplasms , Plants, Medicinal , Drugs, Chinese Herbal/therapeutic use , Humans , Medicine, Chinese Traditional , Neoplasms/drug therapy
11.
Biomed Pharmacother ; 129: 110440, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32768942

ABSTRACT

Jian-pi-yang-zheng Decoction (JPYZ) is a traditional Chinese medicine that is used for the treatment of advanced gastric cancer, and it shows good efficacy in patients. A previous study indicated that JPYZ inhibited the progression of gastric cancer via the regulation of tumor-associated macrophages (TAMs), but the underlying molecular target of JPYZ regulation of TAMs has not been determined. The present study used modified-JPYZ (mJPYZ) to extend our investigation of gastric cancer. Our results showed that mJPYZ inhibited gastric cancer progression in vivo and in vitro. We found that mJPYZ decreased the activity of PI3-kinase γ (PI3Kγ) in TAMs, reduced the anti-inflammatory factor IL-10 and increased the expression of pro-inflammatory cytokines, such as TNF-α and IL-1ß, which ultimately promoted the conversion of TAMs from M2 to M1. Our findings also indicated that mJPYZ inhibited the growth and metastasis of gastric cancer by alleviating the unfavorable differentiation of TAMs via the PI3Kγ signaling cascades. In conclusion, the present findings indicated that mJPYZ inhibited gastric cancer cell EMT via PI3Kγ-dependent TAM reprogramming, which eventually suppressed gastric cancer growth and metastasis. Our study provides an underlying mechanism of a Chinese medicine in the treatment of gastric cancer via PI3Kγ in macrophages.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Drugs, Chinese Herbal/pharmacology , Protein Kinase Inhibitors/pharmacology , Stomach Neoplasms/drug therapy , Tumor-Associated Macrophages/drug effects , Animals , Cell Differentiation/drug effects , Cell Movement/drug effects , Coculture Techniques , Cytokines/metabolism , Disease Progression , Epithelial-Mesenchymal Transition/drug effects , Humans , Inflammation Mediators/metabolism , Mice , Neoplasm Metastasis , Phenotype , Signal Transduction/drug effects , Stomach Neoplasms/enzymology , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , THP-1 Cells , Tumor Microenvironment , Tumor-Associated Macrophages/enzymology , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/pathology
12.
Inorg Chem ; 59(15): 10548-10556, 2020 Aug 03.
Article in English | MEDLINE | ID: mdl-32657582

ABSTRACT

Two types of experiments conducted to investigate the effect of pressure on the spin crossover (SCO) properties of the 2D Fe(II) coordination polymer formulated {Fe[bipy(ttr)2]}n are reported, namely, (1) magnetic measurements performed at variable temperature and at fixed pressure and (2) visible spectroscopy at variable pressure and fixed temperature. The magnetic experiments carried out under a hydrostatic pressure constraint of 0.04, 0.08, and 0.8 GPa reveal a two-step spin transition behavior. The characteristic critical temperatures of the spin transition are shifted upward in temperature as pressure increases. The slope of the straight-line of the Tc vs P plot, dTc/dP, is 775 K/GPa and 300 K/GPa, for the high temperature and the low temperature steps, respectively. These values are remarkably large and denote the extreme sensitivity of the material to the application of pressure. Indeed, the visible spectroscopic measurements performed at 293 K show that a complete spin transition is induced at pressures as low as 0.4 GPa. Moreover, the pressure-induced spin transition is reversible and shows an asymmetric hysteresis. An analysis of the cooperative interactions of the thermal- and pressure-induced spin transition in the framework of the model of elastic interactions reveals that the elastic energy of the lattice as well as the interaction parameter between the SCO centers change during the course of the spin transition. Consequently, the character of the spin transition varies from abrupt for the high temperature step to continuous for the low temperature step.

13.
Pharmacol Res ; 152: 104623, 2020 02.
Article in English | MEDLINE | ID: mdl-31899315

ABSTRACT

Gastric cancer remains the second most common tumor in China. Modified-Bu-zhong-yi-qi decoction (mBYD) as an adjuvant therapy for gastric cancer patients after chemotherapy could significantly prolong the survival time of patients. However, the potential anticancer mechanism for mBYD has not been well characterized. Here, we conducted a comprehensive study of mBYD on a gastric cancer xenograft model with MFC cells in 615 mice and patients. Our results showed that the survival times of the 5-FU + mBYD and mBYD groups were significantly longer than that of the control group. Moreover, the 5-FU + mBYD group had a longer survival time than the 5-FU group. Flow cytometry revealed that the value of CD4+/CD8+ in the mBYD group increased and that the proportions of CD8+PD-1+ T cells and PD-1+Treg cells were decreased when compared to the control group. Compared with the 5-FU group, CD8+PD-1+ T cells and Treg cells were both decreased when 5-FU was combined with mBYD. Further analysis showed that mBYD inhibited PD-L1 expression by the PI3K/AKT pathway in gastric cancer. An in vitro study also showed that mBYD directly promoted the proliferation, activation and cytotoxicity of T lymphocytes. Meanwhile, mBYD reduced the upregulation of CD8+PD-1+ T cells induced by chemotherapy in patients with gastric cancer. In conclusion, mBYD could modulate peripheral immunity and suppress the immune escape of tumors, which may be a promising therapy for gastric cancer.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Drugs, Chinese Herbal/therapeutic use , Fluorouracil/therapeutic use , Stomach Neoplasms/drug therapy , Adenocarcinoma/immunology , Adult , Aged , Animals , Antineoplastic Agents/pharmacology , B7-H1 Antigen/immunology , Cell Line, Tumor , Cell Survival/drug effects , Drug Combinations , Drug Synergism , Drugs, Chinese Herbal/pharmacology , Female , Fluorouracil/pharmacology , Humans , Immunization , Male , Mice , Middle Aged , Programmed Cell Death 1 Receptor/immunology , Stomach Neoplasms/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
14.
BMC Complement Altern Med ; 19(1): 103, 2019 May 10.
Article in English | MEDLINE | ID: mdl-31077221

ABSTRACT

BACKGROUND: Chinese herbal bath therapy (CHBT) is a traditional external therapy that has been used for the treatment of uremic pruritus (UP) in China. We conducted a meta-analysis to evaluate the efficacy and safety of CHBT for UP. METHODS: We searched seven databases for studies published since database inception to September 1, 2018. Randomized trials evaluating CHBT for UP were collected. The therapeutic effects of CHBT were measured by the pruritus level (via the visual analogue scale (VAS) or the symptom score scale) and the total effective rate. We combined studies using mean difference (MD) for continuous outcomes and using risk ratio for dichotomous data, both with 95% confidence intervals. RevMan V.5.3 software was used to assess the data reported and perform the meta-analysis. RESULTS: Seventeen articles including 970 patients were identified. All participants were haemodialysis (HD) patients. CHBT is administered by immersing the whole body in a prepared herbal water bath. On average, an herbal bath prescription included 11 Chinese herbs. The mean treatment duration was 4.7 weeks. Compared with basic treatment (HD or haemoperfusion (HP)) and adding a control of sham CHBT, clear hot water bath, or calamine lotion, CHBT plus basic treatment reduced the VAS score (MD = - 2.38; 95% confidence intervals [CI], - 3.02 to - 1.74; P < 0.00001) and the symptom score (MD = - 8.42; 95% confidence intervals [CI], - 12.47 to - 4.36; P < 0.00001) and had a higher total effectiveness rate (risk ratio [RR] = 1.46; 95% CI, 1.31 to 1.63; P < 0.00001). CONCLUSIONS: In conclusion, CHBT could be a complementary therapy for improving pruritic symptoms in uraemia patients. More rigorously designed, multicentre, prospective RCTs are warranted to further identify the efficacy and safety of CHBT. TRIAL REGISTRATION: Systematic review registration: [PROSPERO registration: CRD42018108506 ].


Subject(s)
Baths , Drugs, Chinese Herbal/therapeutic use , Pruritus/therapy , Uremia/therapy , Humans , Medicine, Chinese Traditional , Randomized Controlled Trials as Topic , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...