Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
1.
Commun Biol ; 7(1): 622, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38783005

ABSTRACT

Recent studies have highlighted the significance of cellular metabolism in the initiation of clonal expansion and effector differentiation of T cells. Upon exposure to antigens, naïve CD4+ T cells undergo metabolic reprogramming to meet their metabolic requirements. However, only few studies have simultaneously evaluated the changes in protein and metabolite levels during T cell differentiation. Our research seeks to fill the gap by conducting a comprehensive analysis of changes in levels of metabolites, including sugars, amino acids, intermediates of the TCA cycle, fatty acids, and lipids. By integrating metabolomics and proteomics data, we discovered that the quantity and composition of cellular lipids underwent significant changes in different effector Th cell subsets. Especially, we found that the sphingolipid biosynthesis pathway was commonly activated in Th1, Th2, Th17, and iTreg cells and that inhibition of this pathway led to the suppression of Th17 and iTreg cells differentiation. Additionally, we discovered that Th17 and iTreg cells enhance glycosphingolipid metabolism, and inhibition of this pathway also results in the suppression of Th17 and iTreg cell generation. These findings demonstrate that the utility of our combined metabolomics and proteomics analysis in furthering the understanding of metabolic transition during Th cell differentiation.


Subject(s)
Cell Differentiation , Metabolomics , Proteomics , Sphingolipids , Sphingolipids/metabolism , Sphingolipids/biosynthesis , Proteomics/methods , Animals , Metabolomics/methods , Mice , Mice, Inbred C57BL
2.
Sci Rep ; 14(1): 1525, 2024 01 17.
Article in English | MEDLINE | ID: mdl-38233537

ABSTRACT

The combination of oncogenes and tumor suppressors is involved in cancer development; however, it is still unknown whether their combination plays a critical role in cancer metastasis. We herein investigated whether genetic combinations affected cell migration ability by establishing the immortalized melanocytes, melan-a cells, with an oncogene, either BRAFV600E or GNA11Q209L, and the loss of mouse Pten. The loss of mouse Pten or human PTEN increased the cell migration ability of our established cells and human melanoma cell lines with oncogenic MAPK signaling and the BRAFV600E or NRASQ61R background, but not with the GNA11Q209L background or no oncogenes. Although increased migration was not related to PI3K-AKT activation, those migration is regulated by the induction of some components in the WAVE regulatory complex, resulting in a higher rate of the formation of lamellipodia. On the other hand, BRAFV600E induced EphA2 phosphorylation at serine 897 through RSK and was also required for cell migration and the formation of lamellipodia. Therefore, the oncogenic MAPK pathway and loss of Pten in melanoma were important for cell migration through the formation of lamellipodia, suggesting the significance of an appropriate combination of genetic alterations not only in cancer development, but also cancer metastasis.


Subject(s)
Melanoma , Animals , Humans , Mice , Cell Line, Tumor , Melanocytes/metabolism , Melanoma/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Pseudopodia/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism
3.
Int Immunol ; 36(3): 129-139, 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38041796

ABSTRACT

To meet the energetic requirements associated with activation, proliferation, and survival, T cells switch their metabolic signatures from energetically quiescent to activated. However, little is known about the role of metabolic pathway controlling the development of invariant natural killer T (iNKT) cells. In the present study, we found that acetyl-CoA carboxylase 1 (ACC1), a rate-limiting enzyme for the fatty acid biosynthesis pathway, plays an essential role in the development of iNKT cells in the thymus. Mice lacking T-cell specific ACC1 showed a reduced number of iNKT cells with an increased proportion of iNKT cells at immature stages 0 and 1. Furthermore, mixed bone marrow (BM) chimera experiments revealed that T-cell intrinsic ACC1 expression was selectively important for the development of thymic iNKT cells, especially for the differentiation of the NKT1 cell subset. Our single-cell RNA-sequencing (scRNA-seq) data and functional analysis demonstrated that ACC1 is responsible for survival of developing iNKT cells. Thus, these findings highlighted a novel role of ACC1 in controlling thymic iNKT cell development mediated by the control of cell survival.


Subject(s)
Natural Killer T-Cells , Mice , Animals , Thymus Gland , Cell Differentiation , Adipogenesis , Fatty Acids/metabolism , Acetyl-CoA Carboxylase/genetics , Acetyl-CoA Carboxylase/metabolism
4.
Sci Immunol ; 8(86): eadd4346, 2023 08 04.
Article in English | MEDLINE | ID: mdl-37540735

ABSTRACT

Metabolic fluxes involving fatty acid biosynthesis play essential roles in controlling the differentiation of T helper 17 (TH17) cells. However, the exact enzymes and lipid metabolites involved, as well as their link to promoting the core gene transcriptional signature required for the differentiation of TH17 cells, remain largely unknown. From a pooled CRISPR-based screen and unbiased lipidomics analyses, we identified that 1-oleoyl-lysophosphatidylethanolamine could act as a lipid modulator of retinoid-related orphan receptor gamma t (RORγt) activity in TH17 cells. In addition, we specified five enzymes, including Gpam, Gpat3, Lplat1, Pla2g12a, and Scd2, suggestive of the requirement of glycerophospholipids with monounsaturated fatty acids being required for the transcription of Il17a. 1-Oleoyl-lysophosphatidylethanolamine was reduced in Pla2g12a-deficient TH17 cells, leading to the abolition of interleukin-17 (IL-17) production and disruption to the core transcriptional program required for the differentiation of TH17 cells. Furthermore, mice with T cell-specific deficiency of Pla2g12a failed to develop disease in an experimental autoimmune encephalomyelitis model of multiple sclerosis. Thus, our data indicate that 1-oleoyl-lysophosphatidylethanolamine is a lipid metabolite that promotes RORγt-induced TH17 cell differentiation and the pathogenicity of TH17 cells.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Nuclear Receptor Subfamily 1, Group F, Member 3 , Mice , Animals , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Cell Differentiation , Lipids
5.
J Biol Chem ; 299(5): 104699, 2023 05.
Article in English | MEDLINE | ID: mdl-37059179

ABSTRACT

The receptor tyrosine kinase ephrin type-A receptor 2 (EphA2) is overexpressed in malignant tumors. We previously reported that non-canonical EphA2 phosphorylation at Ser-897 was catalyzed by p90 ribosomal S6 kinase (RSK) via the MEK-ERK pathway in ligand- and tyrosine kinase-independent manners. Non-canonical EphA2 activation plays a key role in tumor progression; however, its activation mechanism remains unclear. In the present study, we focused on cellular stress signaling as a novel inducer of non-canonical EphA2 activation. p38, instead of ERK in the case of epidermal growth factor signaling, activated RSK-EphA2 under cellular stress conditions, including anisomycin, cisplatin, and high osmotic stress. Notably, p38 activated the RSK-EphA2 axis via downstream MAPK-activated protein kinase 2 (MK2). Furthermore, MK2 directly phosphorylated both RSK1 Ser-380 and RSK2 Ser-386, critical residues for the activation of their N-terminal kinases, which is consistent with the result showing that the C-terminal kinase domain of RSK1 was dispensable for MK2-mediated EphA2 phosphorylation. Moreover, the p38-MK2-RSK-EphA2 axis promoted glioblastoma cell migration induced by temozolomide, a chemotherapeutic agent for the treatment of glioblastoma patients. Collectively, the present results reveal a novel molecular mechanism for non-canonical EphA2 activation under stress conditions in the tumor microenvironment.


Subject(s)
Glioblastoma , Receptor, EphA2 , Signal Transduction , Humans , Anisomycin/pharmacology , Cell Movement , Cisplatin/pharmacology , MAP Kinase Signaling System/physiology , Osmotic Pressure , Phosphorylation , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, EphA2/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Tumor Microenvironment
6.
DNA Res ; 30(1)2023 Feb 01.
Article in English | MEDLINE | ID: mdl-36579714

ABSTRACT

Functionally distinct CD4+ helper T (Th) cell subsets, including Th1, Th2, Th17, and regulatory T cells (Treg), play a pivotal role in the regulation of acquired immunity. Although the key proteins involved in the regulation of Th cell differentiation have already been identified how the proteogenomic landscape changes during the Th cell activation remains unclear. To address this issue, we characterized proteogenomic signatures of differentiation to each Th cell subsets by RNA sequencing and liquid chromatography-assisted mass spectrometry, which enabled us to simultaneously quantify more than 10,000 protein-coding transcripts and 8,000 proteins in a single-shot. The results indicated that T cell receptor activation affected almost half of the transcript and protein levels in a low correlative and gene-specific manner, and specific cytokine treatments modified the transcript and protein profiles in a manner specific to each Th cell subsets: Th17 and Tregs particularly exhibited unique proteogenomic signatures compared to other Th cell subsets. Interestingly, the in-depth proteome data revealed that mRNA profiles alone were not enough to delineate functional changes during Th cell activation, suggesting that the proteogenomic dataset obtained in this study serves as a unique and indispensable data resource for understanding the comprehensive molecular mechanisms underlying effector Th cell differentiation.


Subject(s)
CD4-Positive T-Lymphocytes , Proteogenomics , CD4-Positive T-Lymphocytes/metabolism , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , Cell Differentiation/genetics
7.
Mol Cancer Res ; 21(1): 76-85, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36162957

ABSTRACT

IMPLICATIONS: Considering the importance of GSTA4 in controlling IFNγ responsiveness and the metastatic potential of other melanoma cells, our results highlight a novel mechanism whereby cancer cells escape from host immunity and gain metastatic ability by acquiring resistance to oxidative stress responses through the upregulation of GSTA4.


Subject(s)
Interferon-gamma , Melanoma , Humans , Interferon-gamma/genetics , Melanoma/pathology , Up-Regulation , Neoplasm Metastasis
8.
Biol Pharm Bull ; 45(10): 1553-1558, 2022.
Article in English | MEDLINE | ID: mdl-36184515

ABSTRACT

9,10-Phenanthrenequinone (9,10-PQ), a polycyclic aromatic hydrocarbon that is present in air pollutants, such as diesel exhaust gas and PM2.5, causes the production of excess reactive oxygen species. 9,10-PQ was recently shown to induce the activation of epidermal growth factor receptor (EGFR) by inhibiting protein tyrosine phosphatase 1B. In the present study, we focused on the non-canonical regulation of EGFR, including negative feedback and internalization. In contrast to previous findings, 9,10-PQ inhibited the constitutive tyrosine phosphorylation of EGFR via the mitogen-activated protein extracellular kinase (MEK)/extracellular signal-regulated kinase (ERK)-mediated phosphorylation of Thr-669 in EGFR-overexpressing A431 and MDA-MB-468 cells. In addition, 9,10-PQ induced the clathrin-mediated endocytosis of EGFR via the p38 phosphorylation of Ser-1015 in HeLa and A549 cells. These results revealed that 9,10-PQ strongly induced the non-canonical regulation of EGFR by activating mitogen-activated protein kinase (MAPK).


Subject(s)
Air Pollutants , Phenanthrenes , Air Pollutants/toxicity , Clathrin/metabolism , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogens , Particulate Matter , Phenanthrenes/pharmacology , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Reactive Oxygen Species/metabolism , Tyrosine/metabolism , Vehicle Emissions
9.
Front Immunol ; 13: 904875, 2022.
Article in English | MEDLINE | ID: mdl-36059459

ABSTRACT

Type I interferons (type I-IFN) are critical for the host defense to viral infection, and at the same time, the dysregulation of type I-IFN responses leads to autoinflammation or autoimmunity. Recently, we reported that the decrease in monounsaturated fatty acid caused by the genetic deletion of Scd2 is essential for the activation of type I-IFN signaling in CD4+ Th1 cells. Although interferon regulatory factor (IRF) is a family of homologous proteins that control the transcription of type I-IFN and interferon stimulated genes (ISGs), the member of the IRF family that is responsible for the type I-IFN responses induced by targeting of SCD2 remains unclear. Here, we report that the deletion of Scd2 triggered IRF3 activation for type I-IFN production, resulting in the nuclear translocation of IRF9 to induce ISG transcriptome in Th1 cells. These data led us to hypothesize that IRF9 plays an essential role in the transcriptional regulation of ISGs in Scd2-deleted (sgScd2) Th1 cells. By employing ChIP-seq analyses, we found a substantial percentage of the IRF9 target genes were shared by sgScd2 and IFNß-treated Th1 cells. Importantly, our detailed analyses identify a unique feature of IRF9 binding in sgScd2 Th1 cells that were not observed in IFNß-treated Th1 cells. In addition, our combined analyses of transcriptome and IRF9 ChIP-seq revealed that the autoimmunity related genes, which increase in patient with SLE, were selectively increased in sgScd2 Th1 cells. Thus, our findings provide novel mechanistic insights into the process of fatty acid metabolism that is essential for the type I-IFN response and the activation of the IRF family in CD4+ T cells.


Subject(s)
CD4-Positive T-Lymphocytes , Interferon Regulatory Factors , Interferon Type I , Stearoyl-CoA Desaturase , Antiviral Agents , CD4-Positive T-Lymphocytes/metabolism , Gene Expression Regulation , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Interferon Type I/metabolism , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Stearoyl-CoA Desaturase/genetics , Transcriptome
10.
Sci Rep ; 12(1): 11561, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35798841

ABSTRACT

Monoclonal antibodies targeting the epidermal growth factor receptor (EGFR), including cetuximab and panitumumab, have been used in clinic settings to treat cancer. They have also recently been applied to antibody-drug conjugates (ADCs); however, their clinical efficacy is limited by several issues, including lower internalization efficiency. The binding of cetuximab to the extracellular domain of EGFR suppresses ligand-induced events; therefore, we focus on ligand-independent non-canonical EGFR endocytosis for the delivery of ADCs into cells. Tumor necrosis factor-α (TNF-α) strongly induces the endocytosis of the cetuximab-EGFR complex within 15 min via the p38 phosphorylation of EGFR in a tyrosine kinase-independent manner. A secondary antibody conjugated with saporin, a ribosome-inactivating protein, also undergoes internalization with the complex and enhances its anti-proliferative activity. Anti-cancer agents, including cisplatin and temozolomide, also induce the p38-mediated internalization. The results of the present study demonstrate that synchronous non-canonical EGFR endocytosis may be a feasible strategy for promoting the therapeutic efficacy of EGFR-targeting ADCs in clinical settings.


Subject(s)
Antineoplastic Agents , Immunoconjugates , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cetuximab/chemistry , Cetuximab/pharmacology , Endocytosis , ErbB Receptors/metabolism , Immunoconjugates/pharmacology , Ligands , Pharmaceutical Preparations
11.
Biol Pharm Bull ; 45(2): 162-168, 2022.
Article in English | MEDLINE | ID: mdl-35110502

ABSTRACT

The long-term administration of tamoxifen to estrogen receptor α (ERα)-positive breast cancer patients is an established treatment that reduces mortality and recurrence. However, resistance to tamoxifen and an increased risk of endometrial cancer may occur; therefore, the mechanisms by which tamoxifen causes these adverse effects warrant further study. Tamoxifen has been shown to activate mitogen-activated protein kinase (MAPK) in an ERα-independent manner; therefore, we investigated its effects on the MAPK-mediated non-canonical activation of EphA2, a critical event regulating cell migration. Tamoxifen at slightly higher concentrations induced the rapid phosphorylation of EphA2 at Ser-897 via the MAPK/extracellular signal-regulated kinase (ERK) kinase (MEK)-ERK-ribosomal S6 kinases (RSK) pathway in HeLa cells. In addition, tamoxifen significantly enhanced the migration ability of ERα-negative MDA-MB-231 breast cancer cells in RSK- and EphA2-dependent manners. Phosphorylated EphA2 was internalized and re-localized to the plasma membrane, including lamellipodia, in an RSK-dependent manner. Collectively, the present results provide novel insights into the tumor-promoting activity of tamoxifen.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/metabolism , Receptor, EphA2/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Tamoxifen/pharmacology , Cell Line, Tumor , Cell Movement , Estrogen Receptor alpha , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Phosphorylation , Receptor, EphA2/genetics , Ribosomal Protein S6 Kinases, 90-kDa/genetics
12.
J Exp Med ; 218(12)2021 12 06.
Article in English | MEDLINE | ID: mdl-34813654

ABSTRACT

T cells possess distinguishing effector functions and drive inflammatory disorders. We have previously identified IL-5-producing Th2 cells as the pathogenic population predominantly involved in the pathology of allergic inflammation. However, the cell-intrinsic signaling pathways that control the pathogenic Th2 cell function are still unclear. We herein report the high expression of acetyl-CoA carboxylase 1 (ACC1) in the pathogenic CD4+ T cell population in the lung and skin. The genetic deletion of CD4+ T cell-intrinsic ACC1 dampened eosinophilic and basophilic inflammation in the lung and skin by constraining IL-5 or IL-3 production. Mechanistically, ACC1-dependent fatty acid biosynthesis induces the pathogenic cytokine production of CD4+ T cells via metabolic reprogramming and the availability of acetyl-CoA for epigenetic regulation. We thus identified a distinct phenotype of the pathogenic T cell population in the lung and skin, and ACC1 was shown to be an essential regulator controlling the pathogenic function of these populations to promote type 2 inflammation.


Subject(s)
Acetyl-CoA Carboxylase/metabolism , Drug Eruptions/pathology , Pneumonia/pathology , Th2 Cells/pathology , Acetyl-CoA Carboxylase/genetics , Administration, Topical , Animals , Basophils/metabolism , Basophils/pathology , CD4-Positive T-Lymphocytes/pathology , Calcitriol/analogs & derivatives , Calcitriol/toxicity , Drug Eruptions/drug therapy , Drug Eruptions/genetics , Drug Eruptions/metabolism , Fatty Acids/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Interleukin-3/metabolism , Interleukin-5/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Pneumonia/genetics , Pneumonia/metabolism , Th2 Cells/metabolism
13.
Biol Pharm Bull ; 44(11): 1681-1687, 2021.
Article in English | MEDLINE | ID: mdl-34719645

ABSTRACT

The ligand-induced internalization of epidermal growth factor receptor (EGFR) is generally considered to attenuate downstream signaling via its endosomal degradation. However, the endocytosis of an oncogenic EGFR variant III (EGFRvIII) is impaired, which leads to persistent signaling from the cell surface, thereby promoting the proliferation and survival of glioblastoma multiforme (GBM) cells. Cellular stress triggers the non-canonical endocytosis-recycling of EGFR by p38-mediated phosphorylation. In the present study, we used temozolomide (TMZ), the standard chemotherapeutic agent for the treatment of GBM patients, to examine whether EGFRvIII is controlled by a non-canonical mechanism. TMZ triggered the endocytic trafficking of serine phosphorylated EGFRvIII. Moreover, phosphorylation and endocytosis were abrogated by the selective p38 inhibitor SB203580, but not gefitinib, indicating that EGFRvIII is recruited to p38-mediated non-canonical endocytosis. The combination of TMZ and SB203580 also showed potential inhibitory effects on the proliferation and motility of glioblastoma cells.


Subject(s)
Antineoplastic Agents/pharmacology , Endocytosis/drug effects , ErbB Receptors/metabolism , Glioblastoma/drug therapy , Temozolomide/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Anisomycin/pharmacology , Blotting, Western , Cell Line, Tumor , Electrophoresis, Polyacrylamide Gel , Fluorescent Antibody Technique , Glioblastoma/metabolism , Humans , Phosphorylation/drug effects
14.
Cancer Res ; 81(24): 6131-6141, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34728538

ABSTRACT

Loss-of-function mutations of JAK1/2 impair cancer cell responsiveness to IFNγ and immunogenicity. Therefore, an understanding of compensatory pathways to activate IFNγ signaling in cancer cells is clinically important for the success of immunotherapy. Here we demonstrate that the transcription factor SOX10 hinders immunogenicity of melanoma cells through the IRF4-IRF1 axis. Genetic and pharmacologic approaches revealed that SOX10 repressed IRF1 transcription via direct induction of a negative regulator, IRF4. The SOX10-IRF4-IRF1 axis regulated PD-L1 expression independently of JAK-STAT pathway activity, and suppression of SOX10 increased the efficacy of combination therapy with an anti-PD-1 antibody and histone deacetylase inhibitor against a clinically relevant melanoma model. Thus, the SOX10-IRF4-IRF1 axis serves as a potential target that can bypass JAK-STAT signaling to immunologically warm up melanoma with a "cold" tumor immune microenvironment. SIGNIFICANCE: This study identifies a novel SOX10/IRF4 pathway that regulates noncanonical induction of IRF1 independent of the JAK-STAT pathway and can be targeted to improve the efficacy of anti-PD-1 therapy in melanoma.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Immune Checkpoint Inhibitors/pharmacology , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factors/metabolism , Melanoma/drug therapy , Melanoma/immunology , SOXE Transcription Factors/metabolism , Animals , Apoptosis , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , Drug Therapy, Combination , Gene Expression Regulation, Neoplastic , Humans , Immunotherapy , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factors/genetics , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Inbred C57BL , Prognosis , SOXE Transcription Factors/genetics , Survival Rate , Tumor Cells, Cultured
15.
Cell Rep ; 37(6): 109921, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34758300

ABSTRACT

Regulatory T (Treg) cells are critical for immunological tolerance and immune homeostasis. Treg cells strongly rely on mitochondrial metabolism and show a lower level of glycolysis. However, little is known about the role of lipid metabolism in the regulation of Treg cell homeostasis. Some members of the ACSL family of acyl-coenzyme A (CoA) synthases are expressed in T cells, but their function remains unclear. A combination of RNA-sequencing and proteome analyses shows that Acsbg1, a member of ACSL, is selectively expressed in Treg cells. We show that the genetic deletion of Acsbg1 not only causes mitochondrial dysfunction, but it also dampens other metabolic pathways. The extrinsic supplementation of Acsbg1-deficient Treg cells with oleoyl-CoA restores the phenotype of the Treg metabolic signature. Furthermore, this pathway in ST2+ effector Treg cells enhances immunosuppressive capacity in airway inflammation. Thus, Acsbg1 serves as a metabolic checkpoint governing Treg cell homeostasis and the resolution of lung inflammation.


Subject(s)
Coenzyme A Ligases/metabolism , Energy Metabolism , Lung/enzymology , Mitochondria/enzymology , Pneumonia/enzymology , T-Lymphocytes, Regulatory/enzymology , Animals , Coenzyme A Ligases/genetics , Disease Models, Animal , Fatty Acids/metabolism , Gene Expression Regulation, Enzymologic , Homeostasis , Interleukin-33 , Lung/immunology , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/genetics , Mitochondria/immunology , Organelle Biogenesis , Pneumonia/genetics , Pneumonia/immunology , Signal Transduction , T-Lymphocytes, Regulatory/immunology
16.
Commun Biol ; 4(1): 820, 2021 06 29.
Article in English | MEDLINE | ID: mdl-34188173

ABSTRACT

Host lipid metabolism and viral responses are intimately connected. However, the process by which the acquired immune systems adapts lipid metabolism to meet demands, and whether or not the metabolic rewiring confers a selective advantage to host immunity, remains unclear. Here we show that viral infection attenuates the expression of genes related to lipid metabolism in murine CD4+ T cells, which in turn increases the expression of antiviral genes. Inhibition of the fatty acid synthesis pathway substantially increases the basal expression of antiviral genes via the spontaneous production of type I interferon (IFN). Using a combination of CRISPR/Cas9-mediated genome editing technology and a global lipidomics analysis, we found that the decrease in monounsaturated fatty acid caused by genetic deletion of Scd2 in mice was crucial for the induction of an antiviral response through activation of the cGAS-STING pathway. These findings demonstrate the important relationship between fatty acid biosynthesis and type I IFN responses that enhances the antiviral response.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Fatty Acids, Monounsaturated/metabolism , Interferon Type I/pharmacology , Membrane Proteins/physiology , Nucleotidyltransferases/physiology , Stearoyl-CoA Desaturase/physiology , Virus Diseases/immunology , Animals , Host-Pathogen Interactions , Lipid Metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Signal Transduction , Virus Diseases/metabolism
17.
Biomed Pharmacother ; 140: 111737, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34020249

ABSTRACT

Guggulsterone (GS) [4,17(20)-pregnadiene-3,16-dione], is the main active phytosterol constituent in guggul, the gum resin of Commiphora wightii (Arnott.) Bhand./Commiphora mukul Engl. tree, and is known for its medicinal effects. In this study, we report that GSD-1, a structurally-related synthetic GS derivative, strongly inhibits NF-κB activation induced by TNF-α. GSD-1 prevented the nuclear translocation of p65 through the blockade of IκBα degradation and p65 phosphorylation, and further inhibited the activation of upstream kinases, including transforming growth factor-ß activated kinase 1 (TAK1), IκB kinase (IKK) α, and IKKß. Furthermore, GSD-1 inhibited the cell-intrinsic activation of NF-κB, and exerted its direct anti-cancer and anti-metastatic effects in both murine and human breast cancer cell lines. This study demonstrated GSD-1 to be an attractive compound to target NF-κB activation that has potential for treating breast cancer growth and metastasis.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , NF-kappa B/antagonists & inhibitors , Pregnenediones/pharmacology , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Humans , Mice , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Wound Healing/drug effects
18.
BMC Complement Med Ther ; 21(1): 115, 2021 Apr 09.
Article in English | MEDLINE | ID: mdl-33836731

ABSTRACT

BACKGROUND: Morus alba L. bark has been widely used in traditional medicine for treating several inflammatory diseases, such as hypertension, diabetes mellitus and coughing; however, the molecular mechanisms underlying its anti-inflammatory effects are not well understood. METHODS: We examined the effects of an extract of Morus alba L. bark (MabE) on Toll-like receptor (TLR) ligand-induced activation of RAW264.7 macrophages using a luciferase reporter assay and immunoassays. For the in vivo experiment, we used an imiquimod-induced ear edema model to examine the anti-inflammatory effects of MabE. RESULTS: MabE inhibited the TLR ligand-induced activation of NF-κB in RAW264.7 cells without affecting their viability. Consistent with the inhibition of NF-κB activation, MabE also inhibited the production of IL-6 and IL-1ß from TLR ligand-treated RAW264.7 cells. In vivo MabE treatment inhibited the ear swelling of IMQ-treated mice, in addition to the mRNA expression of IL-17A, IL-1ß and COX-2. The increases in splenic γδT cells in IMQ-treated mice and the production of IL-17A from splenocytes were significantly inhibited by MabE treatment. CONCLUSION: Our study suggests that the anti-inflammatory effects of MabE on the activation of the macrophage cell line RAW246.7 by TLRs and IMQ-induced ear edema are through the inhibition of NF-κB activation and IL-17A-producing γδT cells, respectively.


Subject(s)
Ear Diseases/drug therapy , Edema/drug therapy , Morus , Plant Bark , Toll-Like Receptors/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Edema/chemically induced , Female , Imiquimod/adverse effects , Mice , Phytotherapy
19.
Biol Pharm Bull ; 43(12): 1899-1905, 2020.
Article in English | MEDLINE | ID: mdl-33268707

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3) is considered a potential target for cancer treatment because of its relationship with cellular transformation and tumor initiation and progression. In this study, we aimed to identify a new anti-cancer drug candidate from natural products by targeting STAT3 activity. Using STAT3-luciferase reporter cell line, we screened the chemical library of natural products and found that baicalein, a flavone isolated from the roots of Scutelleria baicalensis, strongly suppressed STAT3 activity in breast cancer cells. Baicalein inhibited STAT3 transcriptional activity and its phosphorylation, and further exhibited anti-proliferative effects in breast cancer cells. Moreover, baicalein suppressed the production of interleukin (IL)-6 and the metastatic potential of breast cancer cells both in vitro and in vivo. Collectively, our study suggests baicalein as an attractive phytochemical compound for reducing metastatic potential of breast cancer cells by regulating STAT3 activity.


Subject(s)
Breast Neoplasms/metabolism , Flavanones/pharmacology , Lung Neoplasms/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Animals , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Female , Flavanones/therapeutic use , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C
20.
Mol Med Rep ; 22(6): 5385-5391, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33173971

ABSTRACT

Accumulating evidence suggests that inflammation is linked to multiple pathological processes and induces cellular and molecular damage through the activation of inflammatory signaling pathways, including the NF­κB pathway. The aim of the present study was to identify natural anti­inflammatory products that can target NF­κB activity, in order to establish a novel therapeutic approach for inflammatory diseases. Using a 4T1 breast cancer cell line that expresses the firefly luciferase gene under the control of an NF­κB response element, 112 natural products were tested for their anti­inflammatory properties. Sohakuhi (Morus alba Linn. bark) extract was observed to strongly suppress NF­κB activity without affecting cell viability. To further examine the anti­inflammatory effect of Sohakuhi, tumor necrosis factor­related apoptosis­inducing ligand (TRAIL)­induced cellular damage of human HaCaT keratinocytes was evaluated. While TRAIL triggered the phosphorylation of the p65 subunit of NF­κB, leading to cellular damage in HaCaT cells, treatment with Sohakuhi extract protected HaCaT cells against TRAIL­induced cellular damage. Moreover, Sohakuhi treatment also upregulated the anti­apoptotic proteins Bcl­xL and Bcl­2. Importantly, through chemical fractionation of Sohakuhi extract, moracin O and P were confirmed to mediate its anti­inflammatory effects. Collectively, the present results indicated that Sohakuhi and moracin may represent potential candidates for the development of novel anti­inflammatory drugs.


Subject(s)
Benzofurans/pharmacology , Animals , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacology , Apoptosis Regulatory Proteins/metabolism , Benzofurans/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Humans , Inflammation/pathology , Keratinocytes/metabolism , Mice , Morus/metabolism , NF-kappa B/drug effects , NF-kappa B/metabolism , Phosphorylation , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...