Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters

Country/Region as subject
Publication year range
1.
BMC Genomics ; 25(1): 443, 2024 May 04.
Article in English | MEDLINE | ID: mdl-38704563

ABSTRACT

BACKGROUND: The transcriptome and metabolome dissection of the skeletal muscle of high- and low- growing individuals from a crossbred population of the indigenous Chongming white goat and the Boer goat were performed to discover the potential functional differentially expressed genes (DEGs) and differential expression metabolites (DEMs). RESULTS: A total of 2812 DEGs were detected in 6 groups at three time stages (3,6,12 Month) in skeletal muscle using the RNA-seq method. A DEGs set containing seven muscle function related genes (TNNT1, TNNC1, TNNI1, MYBPC2, MYL2, MHY7, and CSRP3) was discovered, and their expression tended to increase as goat muscle development progressed. Seven DEGs (TNNT1, FABP3, TPM3, DES, PPP1R27, RCAN1, LMOD2) in the skeletal muscle of goats in the fast-growing and slow-growing groups was verified their expression difference by reverse transcription-quantitative polymerase chain reaction. Further, through the Liquid chromatography-mass spectrometry (LC-MS) approach, a total of 183 DEMs in various groups of the muscle samples and these DEMs such as Queuine and Keto-PGF1α, which demonstrated different abundance between the goat fast-growing group and slow-growing group. Through weighted correlation network analysis (WGCNA), the study correlated the DEGs with the DEMs and identified 4 DEGs modules associated with 18 metabolites. CONCLUSION: This study benefits to dissection candidate genes and regulatory networks related to goat meat production performance, and the joint analysis of transcriptomic and metabolomic data provided insights into the study of goat muscle development.


Subject(s)
Goats , Meat , Muscle, Skeletal , Transcriptome , Animals , Goats/genetics , Goats/metabolism , Muscle, Skeletal/metabolism , Meat/analysis , Metabolomics , Gene Expression Profiling , Metabolome
2.
J Transl Med ; 22(1): 580, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898490

ABSTRACT

The importance of the immune microenvironment in poorly cohesive carcinoma (PCC) has been highlighted due to its limited response rate to conventional therapy and emerging treatment resistance. A combination of clinical cohorts, bioinformatics analyses, and functional/molecular experiments revealed that high infiltration of Interferon Induced Protein with Tetratricopeptide Repeats 1 (IFIT1) + tumor-associated neutrophils (TANs) is a distinguishing feature of PCC patients. Upregulation of IFIT1 + TANs promote migration and invasion of gastric cancer (GC) cell lines (MKN45 and MKN74) and stimulates the growth of cell-derived xenograft models. Besides, by promoting macrophage secreted phosphoprotein 1 (SPP1) expression and facilitating cancer-associated fibroblast and endothelial cell recruitment and activation through TANs, IFIT1 promotes a mesenchymal phenotype, which is associated with a poor prognosis. Importantly, compared to non-PCC (NPCC), PCC tumors is more immunosuppressive. Mechanistically, IFIT1 can be stimulated by IFN-γ and contributes to the expression of Programmed Cell Death 1 Ligand (PDL1) in TANs. We demonstrated in mouse models that IFIT1 + PDL1 + TANs can induce acquired resistance to anti-PD-1 immunotherapy, which may be responsible for the difficulty of PCC patients to benefit from immunotherapy. This work highlights the role of IFIT1 + TANs in mediating the remodeling of the tumor immune microenvironment and immunotherapeutic resistance and introduces IFIT1 + TANs as a promising target for precision therapy of PCC.


Subject(s)
Adaptor Proteins, Signal Transducing , Neutrophils , RNA-Binding Proteins , Humans , Neutrophils/immunology , Neutrophils/metabolism , Animals , RNA-Binding Proteins/metabolism , Cell Line, Tumor , Adaptor Proteins, Signal Transducing/metabolism , Tumor Microenvironment/immunology , Female , B7-H1 Antigen/metabolism , Stomach Neoplasms/pathology , Stomach Neoplasms/immunology , Male , Mice , Drug Resistance, Neoplasm , Cell Movement , Immune Tolerance , Immunosuppression Therapy , Gene Expression Regulation, Neoplastic , Neoplasm Invasiveness , Mice, Nude , Immunotherapy , Middle Aged
3.
J Transl Med ; 22(1): 549, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849852

ABSTRACT

Cellular communication (CC) influences tumor development by mediating intercellular junctions between cells. However, the role and underlying mechanisms of CC in malignant transformation remain unknown. Here, we investigated the spatiotemporal heterogeneity of CC molecular expression during malignant transformation. It was found that although both tight junctions (TJs) and gap junctions (GJs) were involved in maintaining the tumor microenvironment (TME), they exhibited opposite characteristics. Mechanistically, for epithelial cells (parenchymal component), the expression of TJ molecules consistently decreased during normal-cancer transformation and is a potential oncogenic factor. For fibroblasts (mesenchymal component), the expression of GJs consistently increased during normal-cancer transformation and is a potential oncogenic factor. In addition, the molecular profiles of TJs and GJs were used to stratify colorectal cancer (CRC) patients, where subtypes characterized by high GJ levels and low TJ levels exhibited enhanced mesenchymal signals. Importantly, we propose that leiomodin 1 (LMOD1) is biphasic, with features of both TJs and GJs. LMOD1 not only promotes the activation of cancer-associated fibroblasts (CAFs) but also inhibits the Epithelial-mesenchymal transition (EMT) program in cancer cells. In conclusion, these findings demonstrate the molecular heterogeneity of CC and provide new insights into further understanding of TME heterogeneity.


Subject(s)
Cancer-Associated Fibroblasts , Cell Communication , Colorectal Neoplasms , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Tumor Microenvironment , Animals , Humans , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Cell Line, Tumor , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/genetics , Epithelial-Mesenchymal Transition/genetics , Gap Junctions/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Spatio-Temporal Analysis , Tight Junctions/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Autoantigens/genetics , Autoantigens/metabolism
4.
Nature ; 561(7724): 547-550, 2018 09.
Article in English | MEDLINE | ID: mdl-30209395

ABSTRACT

Current models of somatosensory perception emphasize transmission from primary sensory neurons to the spinal cord and on to the brain1-4. Mental influence on perception is largely assumed to occur locally within the brain. Here we investigate whether sensory inflow through the spinal cord undergoes direct top-down control by the cortex. Although the corticospinal tract (CST) is traditionally viewed as a primary motor pathway5, a subset of corticospinal neurons (CSNs) originating in the primary and secondary somatosensory cortex directly innervate the spinal dorsal horn via CST axons. Either reduction in somatosensory CSN activity or transection of the CST in mice selectively impairs behavioural responses to light touch without altering responses to noxious stimuli. Moreover, such CSN manipulation greatly attenuates tactile allodynia in a model of peripheral neuropathic pain. Tactile stimulation activates somatosensory CSNs, and their corticospinal projections facilitate light-touch-evoked activity of cholecystokinin interneurons in the deep dorsal horn. This touch-driven feed-forward spinal-cortical-spinal sensitization loop is important for the recruitment of spinal nociceptive neurons under tactile allodynia. These results reveal direct cortical modulation of normal and pathological tactile sensory processing in the spinal cord and open up opportunities for new treatments for neuropathic pain.


Subject(s)
Neural Pathways/physiopathology , Neuralgia/physiopathology , Pyramidal Tracts/physiopathology , Touch/physiology , Animals , Axons , Cholecystokinin/metabolism , Female , Hindlimb/physiopathology , Hyperalgesia/pathology , Hyperalgesia/physiopathology , Interneurons/metabolism , Male , Mice , Neuralgia/pathology , Nociception/physiology , Pyramidal Tracts/pathology , Somatosensory Cortex/pathology , Somatosensory Cortex/physiopathology , Spinal Cord Dorsal Horn/pathology , Spinal Cord Dorsal Horn/physiopathology
5.
Pharmacol Res ; 188: 106644, 2023 02.
Article in English | MEDLINE | ID: mdl-36603607

ABSTRACT

poorly cohesive (PC) gastric cancer (GC) (PC-GC) is a distinct histological subtype of GC and is defined as a tumor consisting of isolated or small clusters of tumor cells with poorly differentiated and metastatic characteristics. According to multiple studies, PC-GC is intrinsically heterogeneous, with mesenchymal variants being the most aggressive. However, to date, the molecular mechanisms associated with PC-GC are still not fully understood. This study investigated the role of the USP51/ZEB1/ACTA2 axis in promoting GC metastasis. Single-cell sequencing revealed that E-box binding homeobox 1 (ZEB1) expression was significantly increased in a subpopulation of low-adherent cells and was an independent prognostic factor in GC patients. Furthermore, the bulk transcriptome analysis revealed a significant positive correlation between Ubiquitin Specific Peptidase 51 (USP51), ZEB1, and Actin Alpha 2 (ACTA2), and our data further confirmed that all three were highly co-localized in PC-GC tissues. According to the findings of in vitro and in vivo experiments, USP51 was able to maintain ZEB1 expression to promote ACTA2 transcription, thereby activating the mesenchymal phenotype of GC cells and promoting tumor metastasis. Moreover, USP51 could recruit and activate stromal cells, including M2-like macrophages and fibroblasts, through cancer cells. Clinical data suggested that overexpression of USP51 predicts that patients have difficulty benefiting from immunotherapy and is associated with immune-exclusion tumor characteristics. Collectively, the findings of this study shed light on a key mechanism by which elevated USP51 expression induces Epithelial-mesenchymal transition (EMT) in GC cells, hence facilitating GC cell proliferation, survival, and dissemination. In this view, USP51/ZEB1/ACTA2 may serve as a candidate therapeutic target against GC metastasis.


Subject(s)
Stomach Neoplasms , Humans , Stomach Neoplasms/pathology , Actins/metabolism , Cell Line, Tumor , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism , Cell Proliferation/genetics , Cell Movement/genetics , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism
6.
Org Biomol Chem ; 19(37): 8021-8024, 2021 09 29.
Article in English | MEDLINE | ID: mdl-34490432

ABSTRACT

Herein we describe a nickel-catalyzed remote hydrosilylation of unconjugated enones with bulky triphenylsilane. A range of Z-silyl enol ethers are obtained as major isomers due to the process of nickel triggered alkene isomerization. Notably, some specific alkyl silyl enol ethers can be prepared from this protocol, which are not easily accessed by the traditional strategy using a strong base and chlorosilane. This reaction features 100% atom economy, simple reaction conditions, and good yields.

7.
J Biochem Mol Toxicol ; 35(9): e22838, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34273909

ABSTRACT

Colorectal cancer (CRC) is the third most common type of cancer. Here, we studied the inhibitory effect of IRAK1 and IRAK4 as a preventive strategy using a colitis-induced tumorigenesis mouse model. CRC clinical data were obtained from the Gene Expression Omnibus (GEO). An experimental inflammation-dependent CRC model was induced by treatment with azoxymethane (AOM) and then dextran sodium sulfate (DSS) in C57BL/6 mice. Mice were administered an IRAK1/4 inhibitor by intraperitoneal injection at 3 mg/kg twice each week for 9 weeks. The IRAK1/4 inhibitor attenuated histological changes and prevented tumor growth. Tumor-associated proteins, including p65 and Ki-67, were downregulated by the IRAK1/4 inhibitor in AOM/DSS-treated mice. Additionally, IRAK1/4 inhibitor administration effectively decreased the expression of inflammatory cytokines. Furthermore, we observed that IRAK1/4 inhibitor treatment attenuated colitis-induced tumorigenesis by inhibiting epithelial-mesenchymal transition. These observations indicate that inhibition of IRAK1 and IRAK4 may suppress experimental colitis-induced tumorigenesis by inhibiting inflammatory responses and epithelial-mesenchymal transition.


Subject(s)
Carcinogenesis/drug effects , Colitis-Associated Neoplasms/drug therapy , Colitis/drug therapy , Epithelial-Mesenchymal Transition/drug effects , Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Neoplasms, Experimental/drug therapy , Protein Kinase Inhibitors/pharmacology , Animals , Carcinogenesis/chemically induced , Carcinogenesis/metabolism , Colitis/chemically induced , Colitis/enzymology , Colitis-Associated Neoplasms/chemically induced , Colitis-Associated Neoplasms/enzymology , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/enzymology , Interleukin-1 Receptor-Associated Kinases/metabolism , Male , Mice , Neoplasm Proteins/metabolism , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/enzymology
8.
Appl Microbiol Biotechnol ; 105(21-22): 8411-8426, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34617139

ABSTRACT

Major depressive disorder (MDD) is a neurasthenic disease, which is the second-largest burden of disease globally. Increasing studies have revealed that depression is associated with abnormalities in gut microbiota and metabolites. Several species of bacteria have been classified as psychobiotics, which confer mental health benefits through interactions with commensal gut microbiota. Therefore, it is essential to identify new psychobiotics and elucidate their mechanisms in the treatment of depression. This study aims to evaluate the antidepressant effect of Akkermansia muciniphila (AKK) in a mouse model of depression induced by chronic restraint stress (CRS). C57BL/6 male mice were divided into three groups: mice subjected to CRS, mice not subjected to CRS, and mice treated with AKK for 3 weeks. Behavioral tests were performed, and hormone, neurotransmitter, and brain-derived neurotrophic factor (BDNF) levels were measured. Cecal microbiota was analyzed using 16S rRNA gene sequencing, and serum metabolites were detected using untargeted metabolomics. In addition, correlations between altered gut microbiota and metabolites with significant variations in serum associated with AKK ameliorating depression were analyzed using Pearson's correlation coefficient. The results revealed that AKK significantly ameliorated depressive-like behavior and restored abnormal variations in depression-related molecular (corticosterone, dopamine, and BDNF). Moreover, AKK altered chronic stress-induced gut microbial abnormalities. Untargeted metabolomics analysis revealed 23 potential biomarkers in serum that could be associated with the mechanisms underlying CRS-induced depression and the therapeutic effects of AKK. Pearson's correlation coefficient analysis revealed that AKK predominantly upregulated ß-alanyl-3-methyl-L-histidine and edaravone to relieve depression. Furthermore, ß-alanyl-3-methyl-L-histidine and edaravone exhibited the antidepressant phenotype in mice subjected to CRS. In conclusion, the study demonstrated that AKK ameliorates chronic stress-induced depressive symptoms in mice by regulating gut microbiota and metabolites. KEY POINTS: • AKK reduces depressive-like behaviors induced by chronic stress. • AKK regulates the gut microbial structure and metabolomics of serum under the chronic stress. • Antidepressant effect of AKK correlates with the increase of ß-alanyl-3-methyl-l-histidine and edaravone.


Subject(s)
Depressive Disorder, Major , Gastrointestinal Microbiome , Probiotics , Akkermansia , Animals , Male , Mice , Mice, Inbred C57BL , RNA, Ribosomal, 16S/genetics
9.
Med Sci Monit ; 27: e927421, 2021 Jan 29.
Article in English | MEDLINE | ID: mdl-33513128

ABSTRACT

BACKGROUND This study explored the mechanism of action of Ephedrae Herba-Cinnamomi Ramulus couplet medicine (MGCM) at the pharmacological level in the treatment of psoriasis. MATERIAL AND METHODS The active ingredients in MGCM were mined through literature retrieval and the BATMAN-TCM database, and potential targets were predicted. In addition, targets associated with psoriasis were acquired using multiple disease-related databases. Thereafter, an interaction network between candidate MGCM targets and the known psoriasis-associated targets was constructed based on the protein-protein interaction (PPI) data, using the STRING database. Then, the topological parameter degree was determined for mining the core targets for MGCM in the treatment of psoriasis, which also represented the major hubs within the PPI network. In addition, the core networks of targets and ingredients were constructed using Cytoscape software to apply MGCM in the treatment for psoriasis. These core targets were then analyzed for Gene Ontology biological processes and Kyoto Encyclopedia of Genes and Genomes pathway enrichment using OmicShare. RESULTS The ingredient-target core network of MGCM for treating psoriasis was constructed; it contained 52 active ingredients and corresponded to 19 core targets. In addition, based on enrichment analysis, these core targets were majorly enriched for several biological processes (immuno-inflammatory responses, leukocyte differentiation, energy metabolism, angiogenesis, and programmed cell death) together with the relevant pathways (Janus kinase-signal transducer and activator of transcription, toll-like receptors, nuclear factor kappaB, vascular endothelial growth factor, and peroxisome proliferator-activated receptor), thus identifying the possible mechanism of action of MGCM in treating psoriasis. CONCLUSIONS The present network pharmacology study indicated that MGCM alleviates various pathological factors of psoriasis through multiple compounds, multiple targets, and multiple pathways.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Ephedra sinica/metabolism , Psoriasis/drug therapy , Databases, Factual , Databases, Genetic , Drugs, Chinese Herbal/chemistry , Ephedra sinica/chemistry , Gene Ontology , Humans , Medicine, Chinese Traditional/methods , Molecular Docking Simulation/methods , Protein Interaction Maps/drug effects , Signal Transduction/drug effects , Software
10.
Exp Cell Res ; 383(1): 111500, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31306656

ABSTRACT

Oxaliplatin has been widely applied in clinical tumor chemotherapy, the treatment failure of which mainly blames on low susceptibility resulted from intrinsic or acquired drug resistance in tumor cells. Microenvironmental hypoxia is one of the important pathological features of solid tumors, which is closely related to the radiochemotherapy tolerance and poor prognosis. Cinnamaldehyde is extracted from Cinnamomum cassia with inhibiting effect against kinds of tumors. In this study, we demonstrated that hypoxia reduced the sensitivity to oxaliplatin in colorectal cancer (CRC) cells via inducing EMT and stemness. Nonetheless, cinnamaldehyde increased the curative effect of oxaliplatin by promoting apoptosis both in vitro and in vivo. Mechanistically, cinnamaldehyde and oxaliplatin synergistically reversed hypoxia-induced EMT and stemness of CRC cells and suppressed hypoxia-activated Wnt/ß-catenin pathway synergistically. These consequences uncovered the potential therapeutic value of cinnamaldehyde and provided novel ideas on improving the sensitivity of oxaliplatin in CRC therapy.


Subject(s)
Acrolein/analogs & derivatives , Apoptosis/drug effects , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Epithelial-Mesenchymal Transition/drug effects , Neoplastic Stem Cells/drug effects , Oxaliplatin/pharmacology , Acrolein/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Cell Movement , Cell Proliferation , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , Humans , Hypoxia/physiopathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
Biol Chem ; 400(6): 765-776, 2019 05 27.
Article in English | MEDLINE | ID: mdl-30763256

ABSTRACT

The Nm23 gene has been acknowledged to play a crucial role in lung cancer metastasis inhibitory cascades controlled by multiple factors. Low expression or allelic deletion of nm23-H1 is strongly linked to widespread metastasis and poor differentiation of non-small cell lung cancer (NSCLC). In this study, nm23-H1 was down regulated in epithelial-mesenchymal transition (EMT) and stemness enhancement under cobalt chloride (CoCl2)-induced hypoxia in NSCLC cells. Moreover, knocking down of nm23-H1 by shRNA apparently promoted hypoxia induced EMT and stemness, which was entirely suppressed via over expression of nm23-H1. Mechanistically, the Wnt/ß-catenin signaling pathway was found to participate in the nm23-H1-mediated process. Besides, XAV939 prohibited cell EMT and stemness which could be impaired by knocking down of nm23-H1, while stable transfection of nm23-H1 attenuated hypoxia phonotype induced by lithium chloride (LiCl). Generally, our experiment provided evidence that nm23-H1 can reverse hypoxia induced EMT and stemness through the inhibition of the Wnt/ß-catenin pathway, which may furnish a deeper perspective into the better treatment or prognosis for NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Cell Hypoxia , Epithelial-Mesenchymal Transition/genetics , Lung Neoplasms/pathology , NM23 Nucleoside Diphosphate Kinases/genetics , NM23 Nucleoside Diphosphate Kinases/physiology , Neoplastic Stem Cells/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Humans , Lung Neoplasms/metabolism , Wnt Signaling Pathway
12.
J Obstet Gynaecol Res ; 45(12): 2307-2317, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31608558

ABSTRACT

This study aimed to conduct a comprehensive analysis of clinical studies on acupuncture treatment for breast cancer-related lymphedema (BCRL), so as to explore the efficacy and safety of acupuncture treatment and provide evidence for the clinical decision-making. Public databases, mainly including China Academic Journals Full-text Database, Database of Chinese Sci-Tech Journal, Wanfang, PubMed, Embase and the Cochrane Library, from the establishment of databases to December 2018 were searched for randomized controlled trials (RCT) of acupuncture for BCRL. Clinical RCT on the treatment of BCRL with acupuncture combined with drugs or functional exercise were enrolled for the analysis. Bias risk and quality were assessed by two investigators according to the Cochrane Handbook 5.1.0 standard, and the Revman 5.3 software was used for meta-analysis. A total of 13 studies were enrolled, comprising 747 patients (377 in the treatment group and 370 in the control group). The results of meta-analysis showed that acupuncture intervention could improve the total effective rate for the treatment of BCRL (odds ratio = 4.62; 95% confidence interval 2.61-8.17). Recent studies suggest that acupuncture therapy can alleviate the upper limb swelling and improve the subjective pain and discomfort in patients with BCRL, regardless of the control intervention used. However, the number of high-quality RCT is low. Moreover, most of the studies adopted inconsistent efficacy indicators. Hence, additional blinded, large-sample, randomized, well-controlled studies with objective and uniform efficacy indicators are needed, especially in China, to confirm the findings.


Subject(s)
Acupuncture Therapy , Breast Neoplasms/complications , Lymphedema/therapy , Acupuncture Therapy/adverse effects , Female , Humans , Quality Assurance, Health Care , Randomized Controlled Trials as Topic
13.
BMC Complement Altern Med ; 19(1): 86, 2019 Apr 23.
Article in English | MEDLINE | ID: mdl-31014289

ABSTRACT

BACKGROUND: The modified Si-Jun-Zi Decoction (SJZ), a Chinese medicine formula, is clinically used against multiple malignancies including colorectal cancer (CRC). This study aims to evaluate the effect of modified SJZ on CRC liver metastasis and identify the therapeutic mechanisms. METHODS: Human CRC cells with GFP fluorescence were transplanted into Balb/c nude mice spleens. Modified SJZ, 5-fluorouracil or the combined treatment was given for 3 weeks. CRC liver metastasis was measured by fluorescence imaging and plasma cytokines were analyzed. Furthermore, the effects of administration time and doses for the modified SJZ were investigated in nude mice. RESULTS: Modified SJZ could increase the survival rate and reduce CRC liver metastasis in the nude mice model. Plasma GM-CSF level was elevated. Three weeks of treatment with the modified SJZ at the full dose (45 g/kg) could significantly increase the number of macrophages but not neutrophils in the spleen. CONCLUSIONS: These results indicate that modified SJZ can inhibit CRC liver metastasis by activating the innate immune system, providing a complementary and alternative therapy for CRC.


Subject(s)
Antineoplastic Agents , Colonic Neoplasms , Drugs, Chinese Herbal , Liver Neoplasms , Macrophages/drug effects , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , HCT116 Cells , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/prevention & control , Liver Neoplasms/secondary , Mice, Inbred BALB C , Mice, Nude , Xenograft Model Antitumor Assays
14.
Biochem Biophys Res Commun ; 505(2): 485-491, 2018 10 28.
Article in English | MEDLINE | ID: mdl-30268504

ABSTRACT

Adverse side effects of conventional chemotherapy, acquired resistance and fatal tumor metastasis of human colorectal cancer (CRC) are propelling the exploration for novel selective anticarcinogens. Solasodine is a main active component isolated from Solanum incanum L that exhibited a potent stemness and invasion inhibitory effect on human colorectal cancer HCT116 cells. Colony Spheroid formation assay showed that solasodine dose-dependently prohibited HCT116 cell stemness. CD133, CD44, Nanog, Oct-4 and Sox-2 were inhibited by solasodine to reverse stemness and similar mechanism was stimulated in vivo. Transwell and scratch wound assays revealed that solasodine impeded HCT116 cell invasion and migration potential strengthened by TGF-ß1. Moreover, solasodine attenuated TGF-ß1-induced EMT and decreased MMPs while in vivo study showed the same trend. The results of this study implied that solasodine may be a novel therapeutic drug for CRC treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Epithelial-Mesenchymal Transition/drug effects , Solanaceous Alkaloids/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Movement , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Humans , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/genetics , Solanaceous Alkaloids/chemistry , Solanaceous Alkaloids/therapeutic use , Transforming Growth Factor beta1/pharmacology
15.
Cancer Sci ; 108(11): 2248-2264, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28803443

ABSTRACT

Solasodine is a main active component isolated from Solanum incanum L. that performs a wide range of functions containing anti-oxidant, anti-infection, and neurogenesis promotion. In this study, we explored the influence of solasodine on three types of human colorectal cancer (CRC) cell lines. The results show that solasodine prohibited CRC cell proliferation dose- and time-dependently and impeded CRC cell motility by downregulating MMPs. Solasodine was also found to fuel caspase-cascade reaction and increase the ratio between Bax and Bcl-2 so as to induce CRC cell apoptosis. When cells were pretreated with AKT activator (insulin-like growth factor-1) followed by solasodine, the solasodine-induced apoptosis was partially abrogated by insulin-like growth factor-1. Moreover, solasodine hindered tumor development and stimulated similar mechanisms in vivo. In general, our study provides the first evidence that solasodine has a suppressive effect on CRC cells and that this agent may be a novel therapeutic drug for CRC treatment.


Subject(s)
Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Insulin-Like Growth Factor I/administration & dosage , Solanaceous Alkaloids/administration & dosage , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Glycogen Synthase Kinase 3 beta/genetics , Humans , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Signal Transduction/drug effects , bcl-2-Associated X Protein/genetics
16.
Cardiovasc Drugs Ther ; 30(6): 587-598, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27844183

ABSTRACT

PURPOSE: Endoplasmic reticulum (ER) stress contributes to pulmonary artery hypertension (PAH). However, the exact roles of ER stress in right ventricular (RV) dysfunction, which is strongly associated with PAH, are largely unknown. Here, we aimed to explore how ER stress affects RV function in a rat PAH model and evaluated the effects of an ER stress inhibitor on RV dysfunction. METHODS: We examined expression changes of an ER marker: chaperone glucose-regulated protein 78 (GRP78), three ER stress sensor proteins: activating transcription factor 6 (ATF6), inositol-requiring enzyme 1 (IRE1), and protein kinase RNA-like endoplasmic reticulum kinase (PERK), and a key ER stress-induced apoptosis indicator: CCAAT/enhancer-binding protein homologous protein (CHOP), with inflammation indicators: interleukin 6 (IL-6), tumor necrosis factor-α (TNF-α), and matrix metalloproteinases (MMPs) in RV at 3, 7, 14 and 28 days following a single dose of monocrotaline (MCT) injection, with or without a preventive treatment [4-phenylbutyric acid (PBA)]. RV function was evaluated by histological, molecular and echocardiographic analysis. RESULTS: 1) GRP78 protein expression started to increase (1.5 ± 0.06 fold change) at 3d post MCT injection, even before the formation of PAH. 2) ATF6, IRE1, and PERK showed distinctive expression patterns post MCT injection. 3) CHOP expression remained low at day 3 & 7, but significantly increased at day 14 (p < 0.05), along with the peak of RV cardiomyocytes apoptosis. 4) PBA inhibited ER stress and alleviated remodeling and dysfunction in the RV. CONCLUSIONS: The early phase of ER stress might benefit RV function, whereas the extended phase led to RV cardiomyocyte apoptosis and dysfunction. Inhibition of ER stress by PBA during PAH directly improved RV function.


Subject(s)
Endoplasmic Reticulum Stress , Heart Ventricles/physiopathology , Hypertension, Pulmonary/physiopathology , Ventricular Dysfunction, Right/physiopathology , Activating Transcription Factor 6/genetics , Activating Transcription Factor 6/metabolism , Animals , Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Heart Ventricles/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Monocrotaline , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Natriuretic Peptide, Brain/blood , Peptide Fragments/blood , Phenylbutyrates/pharmacology , Phenylbutyrates/therapeutic use , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Ventricular Dysfunction, Right/chemically induced , Ventricular Dysfunction, Right/drug therapy , Ventricular Dysfunction, Right/metabolism , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
17.
BMC Complement Altern Med ; 14: 412, 2014 Oct 24.
Article in English | MEDLINE | ID: mdl-25342273

ABSTRACT

BACKGROUND: Colorectal cancer has become one of the leading cause of cancer morbidity and mortality throughout world. Hederagenin, a derivative of oleanolic acid isolated from the leaves of ivy (Hedera helix L.), has been shown to have potential anti-tumor activity. The study was conducted to evaluate whether hederagenin could induce apoptosis of human colon cancer LoVo cells and explore the possible mechanism. METHODS: MTT assay was used for evaluating cell viability while Annexin V-FITC/PI assay and Hoechst 33342 nuclear stainining were used for the determination of apoptosis and mitochondrial membrane potential. DCFH-DA fluorescence staining and flow cytometry were used to measure ROS generation. Real-time PCR and western blot analysis were performed for apoptosis-related protein expressions. RESULTS: MTT assay showed that hederagenin could significantly inhibit the viability of LoVo cells in a concentration-dependent and time-dependent manner by IC50 of 1.39 µM at 24 h and 1.17 µM at 48 h. The apoptosis ratio was significantly increased to 32.46% and 81.78% by the induction of hederagenin (1 and 2 µM) in Annexin V-FITC/PI assay. Hederagenin could also induce the nuclear changes characteristic of apoptosis by Hoechst 33342 nuclear stainining under fluorescence microscopy. DCFH-DA fluorescence staining and flow cytometry showed that hederagenin could increase significantly ROS generation in LoVo cells. Real-time PCR showed that hederagenin induced the up-regulation of Bax and down-regulation of Bcl-2, Bcl-xL and Survivin. Western blotting analysis showed that hederagenin decreased the expressions of apoptosis-associated proteins Bcl-2, procaspase-9, procaspase-3, and polyADP- ribosepolymerase (PARP) were increased, while the expressions of Bax, caspase-3, caspase-9 were increased. However, there was no significant change on caspase-8. CONCLUSIONS: These results indicated that the disruption of mitochondrial membrane potential might contribute to the apoptosis of hederagenin in LoVo cells. Our findings suggested that hederagenin might be a promising therapeutic candidate for human colon cancer.


Subject(s)
Apoptosis/drug effects , Colonic Neoplasms/physiopathology , Hedera/chemistry , Mitochondria/drug effects , Oleanolic Acid/analogs & derivatives , Plant Extracts/pharmacology , Caspase 3/genetics , Caspase 3/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mitochondria/metabolism , Oleanolic Acid/pharmacology , Plant Leaves/chemistry , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
18.
Zhongguo Zhong Xi Yi Jie He Za Zhi ; 34(2): 224-9, 2014 Feb.
Article in Zh | MEDLINE | ID: mdl-24672950

ABSTRACT

OBJECTIVE: To evaluate the effect of melittin and 5-Fu, DDP, and TXT on human gastric cancer cell line BGC-823 and to primarily explore their possible mechanisms. METHODS: Median effect analysis was employed to determine the interaction between melittin and 5-Fu, DDP, TXT by analyzing the relationship between fraction affected (FA) and the combination index (CI) acquired from the dose-effect curve. Expressions of chemotherapeutic agent-associated genes of BGC-823 cells with or without treatment were measured by real-time fluorescent quantitative PCR. RESULTS: (1) Both melittin and chemotherapeutic agents inhibited the growth of BGC-823. (2) For BGC-823 cells were acted by 5-Fu +melittin, when FA ranged between 0.35-0.75, CI was less than 1. For BGC-823 cells were acted by DDP + melittin, when FA ranged 0.55 or so, CI = 1; when Fa ranged below 0.55, CI was less than 1. For BGC-823 cells were acted by TXT + melittin, CI less than 1 could be seen in the whole interval. (3) After treatment suppressed were the expressions of chemotherapeutic agent-associated genes of BGC-823 cells such as thymidylate synthetase (TS), excision repair cross-complementing gene 1 (ERCC1), breast cancer susceptibility gene 1 (BRCA1), beta-tubulin III (TUBB3), and microtubule-associated protein tau (MAPT). CONCLUSIONS: Melittin had a synergistic effect on the cytotoxicity of chemotherapeutic agents. The possible mechanisms might be associated with down-regulating chemotherapeutic agent-associated genes.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Melitten/pharmacology , Cell Line, Tumor , Drug Synergism , Fluorouracil/pharmacology , Humans
19.
Gastroenterol Res Pract ; 2024: 5591298, 2024.
Article in English | MEDLINE | ID: mdl-38634107

ABSTRACT

Objective: This study is aimed at investigating the expression of Met and YAP in gastric cancer and their impact on clinical prognosis. Methods: Tissue samples and clinical data were collected from 89 patients with gastric cancer. Immunohistochemistry was performed to quantify the expression of Met and YAP using tissue microarray. The correlation between the expressions of Met, YAP, and clinicopathological characteristics of patients was determined using a chi-square test. Survival analysis was conducted using the Kaplan-Meier method, while multivariate survival analysis was performed using the Cox proportional hazard model. Bioinformatics analysis was carried out by downloading chip data from TCGA. Results: The expression levels of both Met and YAP were significantly higher in gastric cancer tissues compared to adjacent tissues (P < 0.001). Met expression showed a positive association with P53 and CD133, whereas YAP expression correlated positively with tumor grade and CD133 (P < 0.05). Pearson's analysis revealed a significant correlation between Met expression and VEGFR as well as CD133, while YAP expression correlated with Ki67 and VEGFR (P < 0.05). Patients with high levels of both Met and YAP exhibited decreased survival time (P < 0.01). Furthermore, Met expression, N stage, and VEGFR were identified as independent risk factors for gastric cancer prognosis (P < 0.05), whereas no such association was observed for YAP expression. Bioinformatics analysis demonstrated a significant correlation between the expressions of Met and YAP; both proteins were highly expressed in gastric cancer patients accompanied by markedly reduced survival time. Conclusion: The expressions of Met and YAP are closely associated with the survival outcomes as well as clinicopathological features in patients with gastric cancer. Moreover, our findings highlight that Met serves as an independent prognostic factor for gastric cancer.

20.
Ann Med Surg (Lond) ; 86(1): 172-189, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38222693

ABSTRACT

Background: Depression is becoming an urgent mental health problem. Si-Ni-San has been widely used to treat depression, yet its underlying pharmacological mechanism is poorly understood. Thus, we aim to explore the antidepressant mechanism of Si-Ni-San by chemical analysis and in-silico methods. Methods: Compounds in Si-Ni-San were determined by ultra-high performance liquid chromatography coupled with quadrupole time-of-flight tandem mass spectrometry (UPLC-Q-TOF-MS/MS). Then, bioactive compounds were obtained from Traditional Chinese Medicines for Systems Pharmacology Database and Analysis Platform and SwissADME, and the potential targets of which were acquired from SwissTargetPrediction. Depression-related targets were collected from GeneCards. The intersection between compound-related targets and depression-related targets were screened out, and the overlapped targets were further performed protein-protein interaction, biological functional and pathway enrichment analysis. Finally, networks of Si-Ni-San against depression were constructed and visualized by Cytoscape. Results: One hundred nineteen compounds in Si-Ni-San were determined, of which 24 bioactive compounds were obtained. Then, 137 overlapped targets of Si-Ni-San against depression were collected. AKT1, PIK3R1, PIK3CA, mTOR, MAPK1 and MAPK8 were the key targets. Furthermore, PI3K-Akt signalling pathway, serotonergic synapse, MAPK signalling pathway and neurotrophin signalling pathway were involved in the antidepressant mechanism of Si-Ni-San. It showed that components like sinensetin, hesperetin, liquiritigenin, naringenin, quercetin, albiflorin and paeoniflorin were the mainly key active compounds for the antidepressant effect of Si-Ni-San. Conclusions: This study demonstrated the key components, key targets and potential pharmacological mechanisms of Si-Ni-San against depression. These results indicate that Si-Ni-San is a promising therapeutic approach for treatment of depression, and may provide evidence for the research and development of drugs for treating depression.

SELECTION OF CITATIONS
SEARCH DETAIL