Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.740
Filter
1.
Immunity ; 55(1): 159-173.e9, 2022 01 11.
Article in English | MEDLINE | ID: mdl-34982959

ABSTRACT

To accommodate the changing needs of the developing brain, microglia must undergo substantial morphological, phenotypic, and functional reprogramming. Here, we examined whether cellular metabolism regulates microglial function during neurodevelopment. Microglial mitochondria bioenergetics correlated with and were functionally coupled to phagocytic activity in the developing brain. Transcriptional profiling of microglia with diverse metabolic profiles revealed an activation signature wherein the interleukin (IL)-33 signaling axis is associated with phagocytic activity. Genetic perturbation of IL-33 or its receptor ST2 led to microglial dystrophy, impaired synaptic function, and behavioral abnormalities. Conditional deletion of Il33 from astrocytes or Il1rl1, encoding ST2, in microglia increased susceptibility to seizures. Mechanistically, IL-33 promoted mitochondrial activity and phagocytosis in an AKT-dependent manner. Mitochondrial metabolism and AKT activity were temporally regulated in vivo. Thus, a microglia-astrocyte circuit mediated by the IL-33-ST2-AKT signaling axis supports microglial metabolic adaptation and phagocytic function during early development, with implications for neurodevelopmental and neuropsychiatric disorders.


Subject(s)
Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/metabolism , Microglia/metabolism , Mitochondria/metabolism , Seizures/immunology , Animals , Behavior, Animal , Disease Susceptibility , Electrical Synapses/metabolism , Energy Metabolism , Humans , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-33/genetics , Mice , Mice, Knockout , Microglia/pathology , Neurogenesis/genetics , Oncogene Protein v-akt/metabolism , Phagocytosis , Signal Transduction
2.
Toxicology ; 468: 153104, 2022 02 28.
Article in English | MEDLINE | ID: mdl-35090964

ABSTRACT

Mycotoxins can impart different types of combined toxicity to humans and animals, therefore, it is critical to understand the underlying mechanisms to eliminate the harm. Herein a combination of zearalenone (ZEA) at 2 µM and deoxynivalenol (DON) at 0.1 µM decreased cell viability and increased ROS level in HepG2 cells, suggesting synergistic toxicity exerted by ZEA and DON even at their low toxic concentrations. Moreover, apoptosis and inflammatory response were promoted after the co-exposure of ZEA and DON, indicated by the increased expression of BAX, Caspase-3, IL-1ß and IL-6 genes. Such synergistic toxicity was closely associated with miR-221-mediated PTEN/PI3K/AKT signal pathway, with a negative regulatory relationship between PTEN and PI3K/AKT signaling. MiR-221 could influence cell viability and ROS level to counter the combined toxicity of ZEA and DON through targeting directly PTEN gene. This study demonstrated the toxicological impact of mycotoxin interactions on cells, and critical role of the interplay between miRNAs and PTEN in monitoring the synergistic toxicity of mycotoxin mixture.


Subject(s)
Hep G2 Cells/drug effects , Oncogene Protein v-akt/metabolism , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Trichothecenes/toxicity , Zearalenone/toxicity , Blotting, Western , Drug Combinations , Drug Synergism , Hep G2 Cells/metabolism , Humans , Inhibitory Concentration 50 , MicroRNAs/metabolism , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction
3.
Biomed Pharmacother ; 146: 112547, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34929579

ABSTRACT

Prolonged exposure to polycyclic aromatic hydrocarbons (PAHs) may result in autoimmune diseases, such as rheumatoid arthritis (RA) and osteoporosis (OP), which are based on an imbalance in bone homeostasis. These diseases are characterized by bone erosion and even a disruption in homeostasis, including in osteoblasts and osteoclasts. Current evidence indicates that multiple factors affect the progression of bone homeostasis, such as genetic susceptibility and epigenetic modifications. However, environmental factors, especially PAHs from various sources, have been shown to play an increasingly prominent role in the progression of bone homeostasis. Hence, it is essential to investigate the effects and pathogenesis of PAHs in bone homeostasis. In this review, recent progress is summarized concerning the effects and mechanisms of PAHs and their ligands and receptors in bone homeostasis. Moreover, strategies based on the effects and mechanisms of PAHs in the regulation of the bone balance and alleviation of bone destruction are also reviewed. We further discuss the future challenges and perspectives regarding the roles of PAHs in autoimmune diseases based on bone homeostasis.


Subject(s)
Bone and Bones/drug effects , Environmental Exposure/adverse effects , Environmental Pollutants/toxicity , Homeostasis/drug effects , Polycyclic Aromatic Hydrocarbons/toxicity , Environmental Exposure/analysis , Environmental Pollutants/analysis , Humans , Mitogen-Activated Protein Kinases/drug effects , NF-kappa B/drug effects , Oncogene Protein v-akt/drug effects , Osteoblasts/drug effects , Osteoclasts/drug effects , Osteoporosis/pathology , Phosphatidylinositol 3-Kinases/drug effects , Polycyclic Aromatic Hydrocarbons/analysis , Signal Transduction/drug effects
4.
Int Immunopharmacol ; 103: 108427, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34922249

ABSTRACT

Preclinical and clinical data show a close relationship between high infiltration of tumor-associated macrophages (TAMs) and a poor prognosis in most types of tumors, thus targeting TAMs stands out as promising anticancer immunotherapies. Recent studies have demonstrated the anti-tumor effects of artemisinin via enhancing anti-tumor immunity within tumor microenvironment, but the underlying mechanism is still not clear. In the present study we uncovered an important role of dihydroartemisinin (DHA) in regulating intratumoral TAM polarization and anti-tumor immune responses in mouse Lewis Lung carcinoma model. We found that DHA inhibited Lewis Lung carcinoma progress, moderately decreased the frequencies of TAMs within tumor stroma, and significantly increased CD86 expression while decreased CD206 expression on TAMs which indicates the role of DHA in polarizing TAMs into a M1-like phenotype. Then, our in vitro data confirmed that DHA dose-dependently promoted macrophage M1 phenotype transition by increasing M1 phenotype-related molecules, meanwhile decreasing the expression of M2 phenotype-related molecules. In addition, DHA increased proinflammatory cytokine production, enhanced the phagocytic capacity while decreased anti-inflammatory cytokine production. Finally, in order to prove that AKT/mTOR signaling potentially mediated DHA-induced macrophage differentiation, we used rapamycin to specifically block the activity of mTOR and stimulated macrophages under M1 stimuli. Our data clearly showed that rapamycin significantly decreased DHA-induced M1-related phenotypes and proinflammatory cytokine expression. In summary, our study highlighted DHA as one of future potential therapeutic options for the development of novel anticancer immunotherapies in lung cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Artemisinins/therapeutic use , Carcinoma, Lewis Lung/drug therapy , Tumor-Associated Macrophages/immunology , Animals , Carcinogenesis , Cell Differentiation , Cytokines/metabolism , Mice , Mice, Inbred C57BL , Oncogene Protein v-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Th1 Cells/immunology , Tumor Cells, Cultured
5.
J Ethnopharmacol ; 282: 114581, 2022 Jan 10.
Article in English | MEDLINE | ID: mdl-34464697

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: The diterpenoids extracted from Euphorbia kansui S.L. Liou ex S.B.Ho, Euphorbia fischeriana Steud. have good antitumor effects. Jolkinolide B has anti-breast cancer effect, but it is unclear whether it has different therapeutic effects between luminal A subtype and luminal B subtype breast cancer. AIM OF THE STUDY: This study investigated the Jolkinolide B has different therapeutic, important targets and pathways effects between luminal A subtype and luminal B subtype breast cancer. MATERIALS AND METHODS: We used bioinformatics to predict the biological process and molecular mechanism of Jolkinolide B in treating two types of breast cancer. Then, in vitro, cultured MCF-7 cells and BT-474 cells were divided into control group, PI3K inhibitor + control group, Jolkinolide B group and PI3K inhibitor + Jolkinolide B group. The CCK-8 assay, Flow cytometric analysis and Transwell cell migration assay was used to detect the cell proliferation, apoptosis, and migration in each group, respectively. ELISA was used to measure the content of Akt and phosphorylated Akt (p-Akt) in cell lysis buffer. RESULTS: Compared to luminal A breast cancer, Jolkinolide B had more targets, proliferation, migration processes and KEGG pathways when treating luminal B subtype breast cancer. Jolkinolide B significantly prolonged the survival time of luminal B subtype breast cancer patients. Compared to the control group, the cell proliferation absorbance value (A value) and migration number of the two kinds of breast cancer cells in the Jolkinolide B group were decreased (P < 0.01, n = 6), and the number of apoptotic cells was increased (P < 0.01, n = 6). Compared to the Jolkinolide B group, the A value and migration number of the two types of breast cancer cells were significantly decreased in the PI3K inhibitor + Jolkinolide B group (P < 0.01, n = 6), and the number of apoptotic cells was significantly increased (P < 0.01, n = 6). In addition, compared to MCF-7 cells, the A value and migration number of BT-474 cells stimulated with Jolkinolide B were significantly decreased (P < 0.01, n = 6), and the number of apoptotic cells was significantly increased (P < 0.01, n = 6). Akt and p-Akt protein levels in the two breast cancer cell lines in the Jolkinolide B group were all decreased (P < 0.01, n = 6), especially in BT-474 cells stimulated by Jolkinolide B. CONCLUSION: Jolkinolide B regulates the luminal A and luminal B subtypes of breast cancer through PI3K-Akt, EGFR and other pathways. Jolkinolide B has more significant therapeutic effect on luminal B subtype breast cancer. In vitro, experiments verified that Jolkinolide B significantly inhibited the proliferation and migration activity of BT-474 breast cancer cells by downregulating the PI3K-Akt pathway.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms , Cell Proliferation/drug effects , Diterpenes/pharmacology , Euphorbia , Signal Transduction/drug effects , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Movement/drug effects , Computational Biology , Down-Regulation , Drugs, Chinese Herbal/pharmacology , Humans , MCF-7 Cells , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Treatment Outcome
6.
Cells ; 10(12)2021 12 06.
Article in English | MEDLINE | ID: mdl-34943938

ABSTRACT

TMEM176B is a member of the membrane spanning 4-domains (MS4) family of transmembrane proteins, and a putative ion channel that is expressed in immune cells and certain cancers. We aimed to understand the role of TMEM176B in cancer cell signaling, gene expression, cell proliferation, and migration in vitro, as well as tumor growth in vivo. We generated breast cancer cell lines with overexpressed and silenced TMEM176B, and a therapeutic antibody targeting TMEM176B. Proliferation and migration assays were performed in vitro, and tumor growth was evaluated in vivo. We performed gene expression and Western blot analyses to identify the most differentially regulated genes and signaling pathways in cells with TMEM176B overexpression and silencing. Silencing TMEM176B or inhibiting it with a therapeutic antibody impaired cell proliferation, while overexpression increased proliferation in vitro. Syngeneic and xenograft tumor studies revealed the attenuated growth of tumors with TMEM176B gene silencing compared with controls. We found that the AKT/mTOR signaling pathway was activated or repressed in cells overexpressing or silenced for TMEM176B, respectively. Overall, our results suggest that TMEM176B expression in breast cancer cells regulates key signaling pathways and genes that contribute to cancer cell growth and progression, and is a potential target for therapeutic antibodies.


Subject(s)
Membrane Proteins/genetics , Oncogene Protein v-akt/genetics , TOR Serine-Threonine Kinases/genetics , Triple Negative Breast Neoplasms/drug therapy , Animals , CD24 Antigen/genetics , CD24 Antigen/immunology , Carcinogenesis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Heterografts , Humans , Mice , RNA-Seq , Signal Transduction/drug effects , Tamoxifen/pharmacology , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/pathology
7.
Int Immunopharmacol ; 101(Pt A): 108312, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34741867

ABSTRACT

Notch signaling regulates the responses of macrophages to different stimuli in a context-dependent manner. The roles of Notch signaling in proinflammatory macrophages are well characterized, whereas its involvement, if any, in IL-4-stimulated macrophages (M(IL-4)) is still unclear. We observed that Notch signaling is functional in human M(IL-4). We performed transcriptome analysis of the Notch1 intracellular domain (NIC1)-overexpressing human monocytic cell line THP-1 with or without IL-4 stimulation to understand the global impact of Notch signaling in M(IL-4). The results revealed that NIC1-overexpressing THP-1 upregulated proinflammatory-associated genes and target genes of IL-4 signaling. We identified serum/glucocorticoid regulated kinase 1 (SGK1) as one of the genes increased by NIC1 overexpression in M(IL-4). To dissect the signaling pathway leading to SGK1 upregulation, we pretreated THP-1-derived macrophages with specific inhibitors of Notch (DAPT), AKT (LY294002) or ERK (U0126). Among these inhibitors, only LY294002 decreased the SGK1 mRNA levels in M(IL-4), indicating that the AKT pathway plays a key role in SGK1 transcription in M(IL-4). Furthermore, treatment of THP-1-derived macrophages with the SGK1 inhibitor (GSK650394) suppressed AKT phosphorylation, but not STAT6, in response to IL-4, indicating that SGK1 positively regulates AKT pathway in M(IL-4). Finally, GSK650394 treatment of human M(IL-4) increased the levels of PPARG mRNA and its protein, indicating a negative role of SGK1 in M(IL-4) function. Overall, we report that the Notch signaling and AKT pathways cooperatively regulate SGK1 expression in M(IL-4) where SGK1, in turn, plays an important role in suppressing IL-4-induced PPARγ expression.


Subject(s)
Immediate-Early Proteins/metabolism , Interleukin-4/pharmacology , Macrophages/metabolism , Oncogene Protein v-akt/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Notch/metabolism , Blotting, Western , Gene Expression Profiling , Humans , Macrophages/drug effects , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects
8.
Neurotox Res ; 39(6): 2007-2017, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34652691

ABSTRACT

Bupivacaine (BUP) is a long-acting amide local anesthetic that may induce strong neurotoxicity and neurological complications. In this study, we elucidate the influence of microRNA-494-3p (miR-494-3p) in BUP-induced neurotoxicity in primary mouse hippocampal neuronal cells. In this study, primary hippocampal neurons were isolated from neonatal C57BL/6 mice. The isolated neurons were treated with various doses of BUP. MTT assay was conducted to analyze neuronal viability. Gene expression measurement was done by RT-qPCR. The impact of miR-494-3p in BUP-mediated neural injury was examined using TUNEL, flow cytometry, western blotting, and ROS activity detection. The regulatory relationship between miR-494-3p and cyclin-dependent kinases 4 and 6 (CDK6) was identified using a luciferase reporter assay. BUP treatment led to neurotoxicity and miR-494-3p upregulation in primary cultured hippocampal neurons. Functionally, miR-494-3p depletion alleviated neuronal apoptosis and oxidative damage induced by BUP. We verified that miR-494-3p targeted and negatively modulated CDK6. MiR-494-3p depletion also activated PI3K/AKT signaling by elevating CDK6 expression in BUP-treated neurons. Furthermore, CDK6 knockdown or PI3K/AKT inactivation attenuated the neuroprotective role of miR-494-3p depletion. Silencing miR-494-3p exerts neuroprotective function in hippocampal neuronal cells against BUP-induced injury by the CDK6-PI3K/AKT pathway.


Subject(s)
Bupivacaine/toxicity , Cyclin-Dependent Kinase 6/metabolism , MicroRNAs/metabolism , Neurotoxicity Syndromes/metabolism , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Animals , Blotting, Western , Flow Cytometry , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Mice , Mice, Inbred C57BL , Neurotoxicity Syndromes/etiology , Reactive Oxygen Species , Reverse Transcriptase Polymerase Chain Reaction
9.
Int Immunopharmacol ; 101(Pt B): 108238, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34688152

ABSTRACT

Senescence marker protein 30 (SMP30) is an aging-related protein that participates in the regulation of tissue damage under various pathological conditions. However, the role of SMP30 in mediating high glucose (HG)-induced injury of retinal ganglion cells (RGCs) has not been fully determined. We found that SMP30 expression declined during HG stimulation in RGCs. Cellular functional studies showed that the up-regulation of SMP30 dramatically prohibited HG-evoked apoptosis, oxidative stress and inflammatory response in RGCs. Mechanism research reported that SMP30 overexpression led to the enhancement of nuclear factor erythroid 2-related factor (Nrf2) activation in HG-stimulated RGCs. Moreover, SMP30 overexpression enhanced the phosphorylation of Akt and glucogen synthase kinase-3ß (GSK-3ß), and the suppression of Akt markedly abolished SMP30-mediated Nrf2 activation in HG-stimulated RGCs. Additionally, the suppression of Nrf2 substantially reversed SMP30-overexpression-induced anti-HG injury effects in RGCs. Overall, these findings suggest that SMP30 protects against HG injury of RGCs by potentiating Nrf2 through regulation of the Akt/GSK-3ß pathway. Our work underscores that SMP30/Akt/GSK-3ß/Nrf2 may exert a vital role in mediating the injury of RGCs during diabetic retinopathy.


Subject(s)
Calcium-Binding Proteins/metabolism , Diabetic Retinopathy/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Retinal Ganglion Cells/physiology , Animals , Apoptosis , Cells, Cultured , Cellular Senescence , Glucose/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Inflammation , Mice , Oncogene Protein v-akt/metabolism , Oxidative Stress , Signal Transduction
10.
Int Immunopharmacol ; 101(Pt B): 108266, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34678694

ABSTRACT

Accumulating evidence has implicated the potential of natural compounds in treatment of asthma. Bixin is a natural food coloring isolated from the seeds of Bixa Orellana, which possesses anti-tumor, anti-inflammatory and antioxidative properties. Nevertheless, its therapeutic effect in asthma has not been elucidated. Our present study demonstrated that administration of Bixin suppressed allergic airway inflammation and reversed glucocorticoids resistance, as well as alleviated airway remodeling and airway hyperresponsiveness (AHR) in asthmatic mice. In vitro studies showed that Bixin treatment could inhibit the development of epithelial-mesenchymal transition (EMT) mediated by transforming growth factor beta (TGF-ß) signaling. Importantly, Bixin antagonized activation of phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) pathway both in vitro and in vivo. Above all, our findings reveal that Bixin functions as a potent antagonist of PI3K/Akt signaling to protect against allergic asthma, highlighting a novel strategy for asthma treatment based on natural products.


Subject(s)
Anti-Asthmatic Agents/therapeutic use , Asthma/drug therapy , Carotenoids/therapeutic use , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Allergens/immunology , Animals , Bixaceae , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred BALB C , Ovalbumin/immunology , Respiratory Hypersensitivity , Signal Transduction , Transforming Growth Factor beta/metabolism
11.
Genes (Basel) ; 12(9)2021 09 11.
Article in English | MEDLINE | ID: mdl-34573384

ABSTRACT

KRAS mutations are one of the most common oncogenic drivers in non-small cell lung cancer (NSCLC) and in lung adenocarcinomas in particular. Development of therapeutics targeting KRAS has been incredibly challenging, prompting indirect inhibition of downstream targets such as MEK and ERK. Such inhibitors, unfortunately, come with limited clinical efficacy, and therefore the demand for developing novel therapeutic strategies remains an urgent need for these patients. Exploring the influence of wild-type (WT) KRAS on druggable targets can uncover new vulnerabilities for the treatment of KRAS mutant lung adenocarcinomas. Using commercially available KRAS mutant lung adenocarcinoma cell lines, we explored the influence of WT KRAS on signaling networks and druggable targets. Expression and/or activation of 183 signaling proteins, most of which are targets of FDA-approved drugs, were captured by reverse-phase protein microarray (RPPA). Selected findings were validated on a cohort of 23 surgical biospecimens using the RPPA. Kinase-driven signatures associated with the presence of the KRAS WT allele were detected along the MAPK and AKT/mTOR signaling pathway and alterations of cell cycle regulators. FoxM1 emerged as a potential vulnerability of tumors retaining the KRAS WT allele both in cell lines and in the clinical samples. Our findings suggest that loss of WT KRAS impacts on signaling events and druggable targets in KRAS mutant lung adenocarcinomas.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Drug Resistance, Neoplasm/genetics , Lung Neoplasms , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins p21(ras)/genetics , A549 Cells , Alleles , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers, Pharmacological/analysis , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Regulatory Networks/drug effects , Gene Regulatory Networks/genetics , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , MTOR Inhibitors/pharmacology , MTOR Inhibitors/therapeutic use , Mutation , Oncogene Protein v-akt/drug effects , Oncogene Protein v-akt/metabolism , Pharmacogenomic Testing , Protein Kinase Inhibitors/pharmacology , Retrospective Studies , Signal Transduction/drug effects , Signal Transduction/genetics
12.
Mol Immunol ; 139: 140-152, 2021 11.
Article in English | MEDLINE | ID: mdl-34509754

ABSTRACT

Neospora caninum is an intracellular parasite which can cause neosporosis and significant economic losses in both dairy and beef industries worldwide. A better understanding of the immune response by host cells against N. caninum could help to design better strategies for the prevention and treatment of neosporosis. Although previous studies have shown TLR2/TLR3 were involved in controlling N. caninum infection in mice, the precise mechanisms of the AKT and MAPK pathways controlled by TLR2/TLR3 to regulate N. caninum-induced IL-12p40 production and the role of TLR2/TLR3 in anti-N. caninum infection in bovine macrophages remain unclear. In the present study, TLR2-/- mice displayed more parasite burden and lower level of IL-12p40 production compared to TLR3-/- mice. N. caninum could activate AKT and ERK signaling pathways in WT mouse macrophages, which were inhibited in TLR2-/- and TLR3-/- mouse macrophages. In N. caninum-infected WT mouse macrophages, AKT inhibitor or AKT siRNA could decrease the phosphorylation of ERK. AKT or ERK inhibitors reduced the production of IL-12p40 and increased the number of parasites. The productions of ROS, NO, and GBP2 were significantly reduced in TLR2-/- and TLR3-/- mouse macrophages. Supplementation of rIL-12p40 inhibited N. caninum proliferation and rescued the productions of IFN-γ, NO, and GBP2 in WT, TLR2-/-, and TLR3-/- mouse macrophages. In bovine macrophages, the expressions of TLR2, TLR3, and IL-12p40 mRNA were significantly enhanced by N. caninum, and N. caninum proliferation was inhibited by TLR2/TLR3 agonists. Taken together, the proliferation of N. caninum in mouse macrophages was controlled by the TLR2/TLR3-AKT-ERK signal pathway via increased IL-12p40 production, which in turn lead to the productions of NO, GBP2, and IFN-γ during N. caninum infection. And in bovine macrophages, TLR2 and TLR3 contributed to inhibiting N. caninum proliferation via increased IL-12p40 production.


Subject(s)
Coccidiosis/immunology , Interleukin-12 Subunit p40/immunology , Macrophages/immunology , Signal Transduction/immunology , Animals , Cattle , MAP Kinase Signaling System/immunology , Mice , Mice, Inbred C57BL , Neospora/immunology , Oncogene Protein v-akt/immunology , Toll-Like Receptor 2/immunology , Toll-Like Receptor 3/immunology
13.
J Immunol ; 207(8): 2179-2191, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34497150

ABSTRACT

Gut microbes play an important role in the development of host B cells. It has been controversial whether GALT is the development site of B cells in pigs. By investigating the relationship between gut microbes and the development of B cells in the GALT of piglets, we found, to our knowledge for the first time, that early B cells exist in the gut lamina propria (LP) in pigs at different ages. We further used Lactobacillus rhamnosus GG (LGG) to treat piglets. The results showed that LGG promotes the development of the early B lineage, affects the composition of the Ig CDR3 repertoires of B cells, and promotes the production of IgA in the intestinal LP. Additionally, we found that the p40 protein derived from LGG can activate the EGFR/AKT and NF-κB signaling pathways, inducing porcine intestinal epithelial cells (IPEC-J2) to secrete a proliferation-inducing ligand (APRIL), which promotes IgA production in B cells. Finally, we identified ARF4 and DIF3 as candidates for p40 receptors on IPEC-J2 by GST pull-down, liquid chromatography-mass spectrometry/mass spectrometry analysis, and coimmunoprecipitation. In conclusion, LGG could promote early B cell differentiation and development in the intestinal LP in piglets and might contribute to promoting IgA production via secretion of p40, which interacts with the membrane receptors on IPEC-J2 and induces them to secrete APRIL. Our study will provide insight to aid in better utilization of probiotics to increase human health.


Subject(s)
B-Lymphocytes/immunology , Bacterial Proteins/metabolism , Gastrointestinal Microbiome/immunology , Immunoglobulin A/metabolism , Intestinal Mucosa/pathology , Lacticaseibacillus rhamnosus/immunology , Mucous Membrane/immunology , Animals , Antibody Formation , Cell Differentiation , Cell Line , Cell Lineage , Green Fluorescent Proteins/metabolism , NF-kappa B/metabolism , Oncogene Protein v-akt/metabolism , Signal Transduction , Swine , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism
14.
Cancer Lett ; 520: 409-421, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34419501

ABSTRACT

Bcl2-associated athanogene 4 (BAG4) has been found to be aberrantly expressed in several types of human cancers. However, little is known about its expression, role, and clinical significance in gastric cancer (GC). In this study, we aimed to address these issues and to explore the underlying mechanisms. The expression level of BAG4, measured by immunohistochemistry, was significantly higher in GC tissues than in paired normal tissues. Elevated BAG4 expression was positively correlated with T stage, lymph node metastasis, and tumor size of GC and was associated with unfavorable outcomes of the patients. The overexpression of BAG4 promoted the in vitro invasion and in vivo metastasis of GC cells, and opposite results were observed after silencing of BAG4. Silencing of BAG4 significantly reduced the phosphorylation of PI3K, AKT, and p65, whereas overexpression of BAG4 markedly enhanced the phosphorylation of these molecules. At the same time, manipulating BAG4 expression resulted in the corresponding changes in p65 nuclear translocation and ZEB1 expression. Luciferase reporter and chromatin immunoprecipitation assays verified that p65 binds to the promoter of ZEB1 to upregulate its transcription. Our results demonstrate that BAG4 plays an oncogenic role in the invasion and metastasis of GC cells by activating the PI3K/AKT/NF-κB/ZEB1 axis to induce epithelial-mesenchymal transition.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Stomach Neoplasms/genetics , Transcription Factor RelA/genetics , Zinc Finger E-box-Binding Homeobox 1/genetics , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , NF-kappa B/genetics , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Oncogene Protein v-akt/genetics , Phosphatidylinositol 3-Kinases/genetics , Signal Transduction/genetics , Stomach Neoplasms/pathology
15.
Exp Mol Pathol ; 122: 104673, 2021 10.
Article in English | MEDLINE | ID: mdl-34371011

ABSTRACT

Recent findings have demonstrated a synergic crosstalk between SHH/GLI and PI3K/Akt/mTORC1 signaling in glioblastoma progression cells in vitro and in tumors in mice, but it is not known if this also occurs in human gliomas. We then aimed to investigate the expression of key proteins of these pathways in different human gliomas. The expression of PTEN, phospho-Akt (Ser473), phospho-S6K1 (Thr389), SHH, GLI1, GLI2 and GLI3 was assessed by immunohistochemistry in gliomas and in control brain tissues. The pattern of expression of each protein was established according to glioma type, glioma grade and to cell type; the relative expression of each protein was used to perform statistical analyses. We found that the expression of proteins of both signaling pathways differs between normal brain and glioma tissues. For instance, normal astrocytes had a different protein expression pattern compared with reactive and tumoral astrocytes. Interestingly, we detected a recurrent pattern of expression of GLI3 in oligodendrocytes and of phospho-S6K1 in mitotic neoplastic cells. We also identified differences of cell signaling according to glioma type: oligodendrogliomas and ependymomas are related with the expression of SHH/GLI proteins. Finally, we detected that high grade gliomas statistically correlate with the expression of GLI1 and GLI2, and that GLI1, GLI2, phospho-Akt and phospho-S6K1 are more expressed in patients with less survival, suggesting that activation of these cell signaling influences glioma outcome and patient survival. In summary, our results show that proteins of PI3K/Akt/mTORC1 and SHH/GLI pathways are differentially expressed in human gliomas according to tumor type and grade, and suggest that the activation of these signaling networks is associated with glioma progression.


Subject(s)
Glioma/genetics , Hedgehog Proteins/genetics , PTEN Phosphohydrolase/genetics , Zinc Finger Protein GLI1/genetics , Adult , Aged , Female , Gene Expression Regulation, Neoplastic/genetics , Glioma/pathology , Humans , Kruppel-Like Transcription Factors/genetics , Male , Mechanistic Target of Rapamycin Complex 1/genetics , Middle Aged , Neoplasm Grading , Oncogene Protein v-akt/genetics , Phosphatidylinositol 3-Kinases/genetics
16.
CNS Neurosci Ther ; 27(11): 1300-1312, 2021 11.
Article in English | MEDLINE | ID: mdl-34346167

ABSTRACT

AIMS: The neurotropic growth factor PDGF-BB was shown to have vital neurorestorative functions in various animal models of Parkinson's disease (PD). Previous studies indicated that the regenerative property of PDGF-BB contributes to the increased intensity of tyrosine hydroxylase (TH) fibers in vivo. However, whether PDGF-BB directly modulates the expression of TH, and the underlying mechanism is still unknown. We will carefully examine this in our current study. METHOD: MPTP-lesion mice received PDGF-BB treatment via intracerebroventricular (i.c.v) administration, and the expression of TH in different brain regions was assessed by RT-PCR, Western blot, and immunohistochemistry staining. The molecular mechanisms of PDGF-BB-mediated TH upregulation were examined by RT-PCR, Western blot, ChIP assay, luciferase reporter assay, and immunocytochemistry. RESULTS: We validated a reversal expression of TH in MPTP-lesion mice upon i.c.v administration of PDGF-BB for seven days. Similar effects of PDGF-BB-mediated TH upregulation were also observed in MPP+ -treated primary neuronal culture and dopaminergic neuronal cell line SH-SY5Y cells. We next demonstrated that PDGF-BB rapidly activated the pro-survival PI3K/Akt and MAPK/ERK signaling pathways, as well as the downstream CREB in SH-SY5Y cells. We further confirmed the significant induction of p-CREB in PDGF-BB-treated animals in vivo. Using a genetic approach, we demonstrated that the transcription factor CREB is critical for PDGF-BB-mediated TH expression. The activation and nucleus translocation of CREB were promoted in PDGF-BB-treated SH-SY5Y cells, and the enrichment of CREB on the promoter region of TH gene was also increased upon PDGF-BB treatment. CONCLUSION: Our data demonstrated that PDGF-BB directly regulated the expression of TH via activating the downstream Akt/ERK/CREB signaling pathways. Our finding will further support the therapeutic potential of PDGF-BB in PD, and provide the possibility that targeting PDGF signaling can be harnessed as an adjunctive therapy in PD in the future.


Subject(s)
Becaplermin/pharmacology , Dopaminergic Neurons/drug effects , Neuroprotective Agents/pharmacology , Signal Transduction/drug effects , Tyrosine 3-Monooxygenase/biosynthesis , Animals , Becaplermin/administration & dosage , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein/drug effects , Female , Humans , Immunohistochemistry , Injections, Intraventricular , MAP Kinase Signaling System/drug effects , MPTP Poisoning/pathology , Mice , Mice, Inbred C57BL , Neuroprotective Agents/administration & dosage , Oncogene Protein v-akt/genetics , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/pathology , Pregnancy
17.
Cancer Sci ; 112(10): 4166-4175, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34288272

ABSTRACT

Various molecular-targeting drugs have markedly improved the treatment of patients with breast cancer. As yet, therapies for triple-negative breast cancer are mainly cytotoxic agents. To investigate the novel therapy for triple-negative breast cancer, we herein examined the effects of a new combination therapy comprising a RAF/MEK inhibitor CH5126766, also known as VS-6766, which we originally discovered, and eribulin. The combination of CH5126766 and eribulin potently inhibited cell growth in the triple-negative breast cancer cell lines tested. The underlying mechanism in the efficacy of this combination treatment in vitro and in vivo was due to enhanced apoptosis through the suppression of survivin and Bcl-2 family proteins. We also showed the suppressed expression of programmed cell death ligand 1 (PD-L1) in combination therapy in vivo. We found that combination therapy with eribulin and CH5126766 for triple-negative breast cancer inhibited cell growth by apoptosis and raised a possibility that immune responses through suppression of PD-L1 might partially contribute to inhibition of tumor growth, indicating the potential of this combination as a novel strategy for triple-negative breast cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Coumarins/therapeutic use , Furans/therapeutic use , Ketones/therapeutic use , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Animals , Apoptosis/drug effects , B7-H1 Antigen/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Mice , Mice, Inbred BALB C , Oncogene Protein v-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Random Allocation , Survivin/metabolism , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/metabolism , Tumor Stem Cell Assay
18.
Genes (Basel) ; 12(7)2021 06 28.
Article in English | MEDLINE | ID: mdl-34203273

ABSTRACT

Circular RNAs (circRNAs), a class of covalently closed RNAs formed by a back-splicing reaction, have been involved in the regulation of diverse oncogenic processes. In this article we describe circVAMP3, a novel circular RNA overexpressed in RH4, a representative cell line of alveolar rhabdomyosarcoma. We demonstrated that circVAMP3 has a differential m6A pattern opposed to its linear counterpart, suggesting that the two isoforms can be differently regulated by such RNA modification. Moreover, we show how circVAMP3 depletion in alveolar rhabdomyosarcoma cells can impair cell cycle progression, through the alteration of the AKT-related pathways, pointing to this non-coding RNA as a novel regulator of the alveolar rhabdomyosarcoma progression and as a putative future therapeutic target.


Subject(s)
Cell Cycle/genetics , RNA, Circular/genetics , RNA/genetics , Rhabdomyosarcoma, Alveolar/genetics , Cell Division/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/genetics , Humans , Myoblasts/metabolism , Oncogene Protein v-akt/genetics , RNA Splicing/genetics , Rhabdomyosarcoma, Alveolar/pathology
19.
Int Immunopharmacol ; 99: 107986, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34303280

ABSTRACT

Albiziae Cortex (AC) is a well-known traditional Chinese medicine with sedative-hypnotic effects and neuroprotective ability. However, the bioactive components of AC responsible for the neuro-protective actitivity remain unknown. Here, we investigated the anti-neuroinflammatory effects of (-)-syringaresinol (SYR) extracted from AC in microglia cells and wild-type mice. As a result, (-)-SYR significantly reduced lipopolysaccharide (LPS)-induced production of interleukin - 6 (IL-6), tumor necrosis factor α (TNF-α), interleukin -1 beta (IL-1ß), cycloxygenase-2 (COX-2), and nitric oxide (NO) in BV2 microglia cells. (-)-SYR also significantly reduced M1 marker CD40 expression and increased M2 marker CD206 expression. Moreover, we found that (-)-SYR inhibited LPS-induced NF-κB activation by suppressing the translocation of NF-κB p65 into the nucleus in a concentration-dependent manner. Meanwhile, estrogen receptor ß (ERß) was found to be implied in the anti-inflammatory activity of (-)-SYR in BV2 microglia. In vivo experiments revealed that administration of (-)-SYR in mice significantly reduced microglia/astrocytes activation and mRNA levels of proinflammatory mediators. Taken together, our data indicated that (-)-SYR exerted the anti-neuroinflammatory effects by inhibiting NF-κB activation and modulation of microglia polarization, and via interaction with ERß. The anti-neuroinflammatory activity of (-)-SYR may provide a new therapeutic avenue for the treatment of brain diseases associated with inflammation.


Subject(s)
Estrogen Receptor beta/metabolism , Furans/pharmacology , Lignans/pharmacology , Microglia/metabolism , Albizzia/chemistry , Animals , Anti-Inflammatory Agents/therapeutic use , Cell Survival/drug effects , Cytokines/metabolism , Estrogen Receptor beta/antagonists & inhibitors , Furans/chemistry , Lignans/chemistry , Lipopolysaccharides/pharmacology , MAP Kinase Signaling System/drug effects , Macrophage Activation/drug effects , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Neuroinflammatory Diseases/drug therapy , Oncogene Protein v-akt/metabolism , Transcription Factor RelA/metabolism
20.
Antiviral Res ; 194: 105145, 2021 10.
Article in English | MEDLINE | ID: mdl-34303747

ABSTRACT

Genital herpes infections in humans are usually caused by herpes simplex virus type-2 (HSV-2), which result in recurrent lesions in the anogenital region. Past studies have shown that a viral protein translation inhibitor, BX795 is capable of mitigating HSV-2 infection both in vitro and in vivo when dosed therapeutically. However, any preventative benefits of this compound against HSV-2 infection remain poorly understood. In this study, we show that BX795 when added prophylactically to human vaginal keratinocytes generates strong preventative effects against a future HSV-2 infection. As a possible mechanism for this action, we found that BX795 efficiently reduces phosphorylation of AKT and its downstream targets p70S6K and 4EBP1. Our in-silico protein docking studies support our immunoblotting results and provide further credence to the proposed mechanism. Using a murine model of vaginal infection, we show that prior treatment with BX795 is also protective in vivo and leads to lower viral replication in the vaginal tissue.


Subject(s)
Antiviral Agents/pharmacology , Keratinocytes/drug effects , Keratinocytes/virology , Oncogene Protein v-akt/antagonists & inhibitors , Pyrimidines/pharmacology , Thiophenes/pharmacology , Vagina/virology , Virus Replication/drug effects , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Animals , Cell Cycle Proteins/antagonists & inhibitors , Female , Herpes Genitalis/prevention & control , Metabolic Networks and Pathways/drug effects , Mice , Mice, Inbred BALB C , Ribosomal Protein S6 Kinases, 70-kDa/antagonists & inhibitors , Vagina/cytology
SELECTION OF CITATIONS
SEARCH DETAIL