Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 704
Filter
1.
Int J Biol Sci ; 20(12): 4635-4653, 2024.
Article in English | MEDLINE | ID: mdl-39309439

ABSTRACT

Darolutamide, an androgen receptor inhibitor, has been approved by the Food and Drug Administration (FDA) for the treatment of prostate cancer (PCa), especially for patients with androgen receptor mutations. Owing to the unique lipidomic profile of PCa and the effect of darolutamide, the relationship between darolutamide and ferroptosis remains unclear. The present study showed that darolutamide significantly induces ferroptosis in AR+ PCa cells. Mechanistically, darolutamide promotes ferroptosis by downregulating SREBP1, which then inhibits the transcription of FASN. FASN knockdown modulates phospholipid remodeling by disrupting the balance between polyunsaturated fatty acids (PUFAs) and saturated fatty acids (SFAs), which induces ferroptosis. Clinically, SREBP1 and FASN are significantly overexpressed in PCa tissues and are related to poor prognosis. Moreover, the synergistic antitumor effect of combination therapy with darolutamide and ferroptosis inducers (FINs) was confirmed in PCa organoids and a mouse xenografts model. Overall, these findings revealed a novel mechanism of darolutamide mediated ferroptosis in PCa, laying the foundation for the combination of darolutamide and FINs as a new therapeutic strategy for PCa patients.


Subject(s)
Fatty Acid Synthase, Type I , Ferroptosis , Prostatic Neoplasms , Sterol Regulatory Element Binding Protein 1 , Ferroptosis/drug effects , Male , Humans , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Animals , Mice , Cell Line, Tumor , Fatty Acid Synthase, Type I/metabolism , Fatty Acid Synthase, Type I/genetics , Phospholipids/metabolism , Pyrazoles/pharmacology , Mice, Nude , Xenograft Model Antitumor Assays
2.
Int Immunopharmacol ; 142(Pt B): 113136, 2024 Dec 05.
Article in English | MEDLINE | ID: mdl-39293316

ABSTRACT

Hyper-activations of monocytes/macrophages and dendritic cells (DCs) contribute to the pathogenesis of various autoimmune diseases, such as systemic lupus erythematosus (SLE). Fatty acid synthase (FASN) is essential for the de novo synthesis of long-chain fatty acids, which play a key role in controlling the activation, differentiation, and function of immune cells. However, the role of FASN in regulating the activations of monocytes/macrophages and DCs has not been studied. In this study, we investigated the involvement of the FASN in modulating the activations of macrophages and DCs, as well as the pathogenesis of SLE. Importantly, we observed a significant upregulation of FASN expression in monocytes and DCs from patients with SLE. This increase is strongly correlated with disease severity and activation status of the immune cells. Furthermore, overexpression of FASN significantly boosts the TLR4/7/9-mediated activation of macrophages and DCs, while knockdown of FASN markedly inhibits this activation. Notably, knockdown of FASN alleviates TLR7 agonist imiquimod (IMQ)-induced lupus in mice and the activation of macrophages and DCs. It makes more sense that pharmaceutical targeting of FASN by using TVB-2640 significantly alleviates IMQ-induced lupus in mice and the activation of macrophages and DCs, as well as in spontaneous lupus MRL/lpr mice. Thus, FASN contributes to the TLRs-mediated activation of macrophages and DCs, as well as the pathogenesis of SLE. More importantly, FASN inhibitor TVB-2640 is expected to be an effective drug in the treatment of SLE.


Subject(s)
Dendritic Cells , Fatty Acid Synthase, Type I , Lupus Erythematosus, Systemic , Macrophages , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Humans , Macrophages/immunology , Macrophages/metabolism , Macrophages/drug effects , Female , Mice , Fatty Acid Synthase, Type I/metabolism , Fatty Acid Synthase, Type I/genetics , Imiquimod , Adult , Male , Toll-Like Receptors/metabolism , Cells, Cultured , Macrophage Activation , Disease Models, Animal
3.
Cancer Biol Ther ; 25(1): 2403197, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-39345091

ABSTRACT

Drug resistance is a critical impediment to efficient therapy of diffuse large B-cell lymphoma (DLBCL) patients. Recent studies have highlighted the association between ferroptosis and drug resistance that has been reported. Fatty acid synthase (FASN) is always related to a poor prognosis. In this study, we investigate the impact of FASN on drug resistance in DLBCL and explore its potential modulation of ferroptosis mechanisms. The clinical correlation of FASN mRNA expression was first analyzed to confirm the role of FASN on drug resistance in DLBCL based on the TCGA database. Next, the impact of FASN on ferroptosis was investigated in vitro and in vivo. Furthermore, a combination of RNA-seq, western blot, luciferase reporter, and ChIP experiments was employed to elucidate the underlying mechanism. The prognosis for patients with DLBCL was worse when FASN was highly expressed, particularly in those undergoing chemotherapy for Adriamycin (ADM). FASN promoted tumor growth and resistance of DLBCL to ADM, both in vitro and in vivo. It is noteworthy that this effect was achieved by inhibiting ferroptosis, since Fer-1 (a ferroptosis inhibitor) treatment significantly recovered the effects of silencing FASN on inhibiting ferroptosis, while Erastin (a ferroptosis inducer) treatment attenuated the impact of overexpressing FASN. Mechanistically, FASN activated NF-κB/STAT3 signaling pathway through phosphorylating the upstream IKKα and IκBα, and the activated STAT3 promoted GPX4 expression by directly binding to GPX4 promoter. FASN inhibits ferroptosis in DLBCL via NF-κB/STAT3/GPX4 signaling pathway, indicating its critical role in mediating ADM resistance of DLBCL.


Subject(s)
Doxorubicin , Drug Resistance, Neoplasm , Fatty Acid Synthase, Type I , Ferroptosis , Lymphoma, Large B-Cell, Diffuse , NF-kappa B , Phospholipid Hydroperoxide Glutathione Peroxidase , STAT3 Transcription Factor , Humans , Ferroptosis/drug effects , Ferroptosis/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoma, Large B-Cell, Diffuse/drug therapy , NF-kappa B/metabolism , Mice , Animals , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Drug Resistance, Neoplasm/genetics , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Fatty Acid Synthase, Type I/metabolism , Fatty Acid Synthase, Type I/genetics , STAT3 Transcription Factor/metabolism , Cell Line, Tumor , Xenograft Model Antitumor Assays , Gene Expression Regulation, Neoplastic/drug effects , Female , Male , Signal Transduction/drug effects , Mice, Nude , Prognosis
4.
Hepatol Commun ; 8(9)2024 09 01.
Article in English | MEDLINE | ID: mdl-39185911

ABSTRACT

BACKGROUND: Dysregulated fatty acid metabolism is closely linked to the development of alcohol-associated liver disease (ALD). KCs, which are resident macrophages in the liver, play a critical role in ALD pathogenesis. However, the effect of alcohol on fatty acid metabolism in KCs remains poorly understood. The current study aims to investigate fatty acid metabolism in KCs and its potential effect on ALD development. METHODS: Wild-type C57BL/6 mice were fed a Lieber-DeCarli ethanol liquid diet for 3 days. Then, the liver injury and levels of intrahepatic bacteria were assessed. Next, we investigated the effects and underlying mechanisms of ethanol exposure on fatty acid metabolism and the phagocytosis of KCs, both in vivo and in vitro. Finally, we generated KCs-specific Fasn knockout and overexpression mice to evaluate the impact of FASN on the phagocytosis of KCs and ethanol-induced liver injury. RESULTS: Using Bodipy493/503 to stain intracellular neutral lipids, we found significantly reduced lipid levels in KCs from mice fed an alcohol-containing diet for 3 days and in RAW264.7 macrophages exposed to ethanol. Mechanistically, alcohol exposure suppressed sterol regulatory element-binding protein 1 transcriptional activity, thereby inhibiting fatty acid synthase (FASN)-mediated de novo lipogenesis in macrophages both in vitro and in vivo. We show that genetic ablation and pharmacologic inhibition of FASN significantly impaired KC's ability to take up and eliminate bacteria. Conversely, KCs-specific Fasn overexpression reverses the impairment of macrophage phagocytosis caused by alcohol exposure. We also revealed that KCs-specific Fasn knockout augmented KCs apoptosis and exacerbated liver injury in mice fed an alcohol-containing diet for 3 days. CONCLUSIONS: Our findings indicate the crucial role of de novo lipogenesis in maintaining effective KCs phagocytosis and suggest a therapeutic target for ALD based on fatty acid synthesis in KCs.


Subject(s)
Fatty Acids , Kupffer Cells , Liver Diseases, Alcoholic , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis , Animals , Kupffer Cells/metabolism , Mice , Fatty Acids/metabolism , Fatty Acids/biosynthesis , Liver Diseases, Alcoholic/metabolism , Ethanol , Fatty Acid Synthase, Type I/metabolism , Fatty Acid Synthase, Type I/genetics , Male , Disease Progression , Liver/metabolism , Lipogenesis/drug effects , RAW 264.7 Cells , Sterol Regulatory Element Binding Protein 1/metabolism , Disease Models, Animal
5.
JCI Insight ; 9(18)2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39115936

ABSTRACT

The interleukin 17 (IL-17) family of cytokines has emerged as a critical player in autoimmune disease, including systemic lupus erythematosus (SLE). However, the role of IL-17B, a poorly understood cytokine, in the pathogenesis of SLE is still not known. In this study, we investigated the role of IL-17B in the activation and differentiation of B cells, and the pathogenesis of SLE. Intriguingly, IL-17B deficiency aggravated disease in lupus-prone mice and promoted the activation of B cells and the differentiation of germinal center B cells and plasma cells, while recombinant mouse IL-17B (rmIL-17B) significantly alleviated disease in lupus-prone mice. Mechanistically, rmIL-17B inhibited the activation of the Toll-like receptor and interferon pathways in B cells by downregulating fatty acid synthase-mediated (FASN-mediated) lipid metabolism. Loss of FASN significantly alleviated the disease in lupus-prone mice and inhibited the activation and differentiation of B cells. In addition, B cells had greater FASN expression and lower IL-17RB levels in patients with SLE than in healthy controls. Our study describes the role of IL-17B in regulating B cell activation and differentiation, and alleviating the onset of SLE. These findings will lay a theoretical foundation for further understanding of the pathogenesis of SLE.


Subject(s)
B-Lymphocytes , Cell Differentiation , Interleukin-17 , Lupus Erythematosus, Systemic , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Animals , Mice , Interleukin-17/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Humans , Female , Fatty Acid Synthase, Type I/metabolism , Fatty Acid Synthase, Type I/genetics , Disease Models, Animal , Male , Lymphocyte Activation/immunology , Mice, Knockout , Lipid Metabolism
6.
Cell Rep ; 43(9): 114681, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39180751

ABSTRACT

Regulatory T cells (Tregs) suppress pro-inflammatory conventional T cell (Tconv) responses. As lipids impact cell signaling and function, we compare the lipid composition of CD4+ thymus-derived (t)Tregs and Tconvs. Lipidomics reveal constitutive enrichment of neutral lipids in Tconvs and phospholipids in tTregs. TNFR2-co-stimulated effector tTregs and Tconvs are both glycolytic, but only in tTregs are glycolysis and the tricarboxylic acid (TCA) cycle linked to a boost in fatty acid (FA) synthesis (FAS), supported by relevant gene expression. FA chains in tTregs are longer and more unsaturated than in Tconvs. In contrast to Tconvs, tTregs effectively use either lactate or glucose for FAS and rely on this process for proliferation. FASN and SCD1, enzymes responsible for FAS and FA desaturation, prove essential for the ability of tTregs to suppress Tconvs. These data illuminate how effector tTregs can thrive in inflamed or cancerous tissues with limiting glucose but abundant lactate levels.


Subject(s)
Fatty Acids , Glucose , Lactic Acid , Stearoyl-CoA Desaturase , T-Lymphocytes, Regulatory , Humans , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Glucose/metabolism , Fatty Acids/metabolism , Lactic Acid/metabolism , Lactic Acid/biosynthesis , Stearoyl-CoA Desaturase/metabolism , Glycolysis , Thymus Gland/metabolism , Thymus Gland/immunology , Fatty Acid Synthase, Type I/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Citric Acid Cycle
7.
Cell Death Dis ; 15(7): 537, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39075049

ABSTRACT

It has been shown that the formation of filopodia is a key step in tumor cell metastasis, but there is limited research regarding its mechanism. In this study, we demonstrated that fatty acid synthase (FASN) promoted filopodia formation in liver cancer cells by regulating fascin actin-bundling protein 1 (FSCN1), a marker protein for filopodia. Mechanistically, on the one hand, the accumulation of FASN is caused by the enhanced deubiquitination of FASN mediated by UCHL5 (ubiquitin c-terminal hydrolase L5). In this pathway, low expression of SIAH1 (Seven in absentia homolog 1) can decrease the ubiquitination and degradation of ADRM1 (adhesion regulating molecule 1) thereby increasing its protein level, which will recruit and activate the deubiquitination enzyme UCHL5, leading to FASN undergo deubiquitination and escape from proteasomal degradation. On the other hand, the accumulation of FASN is related to its weakened ubiquitination, where SIAH1 directly acts as a ubiquitin ligase toward FASN, and low expression of SIAH1 reduces the ubiquitination and degradation of FASN. Both the two pathways are involved in the regulation of FASN in liver cancer. Our results reveal a novel mechanism for FASN accumulation due to the low expression of SIAH1 in human liver cancer and suggest an important role of FASN in filopodia formation in liver cancer cells.


Subject(s)
Liver Neoplasms , Microfilament Proteins , Nuclear Proteins , Pseudopodia , Ubiquitin-Protein Ligases , Ubiquitination , Humans , Pseudopodia/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Microfilament Proteins/metabolism , Microfilament Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Carrier Proteins/metabolism , Carrier Proteins/genetics , Animals , Cell Line, Tumor , Mice, Nude , Fatty Acid Synthase, Type I/metabolism , Fatty Acid Synthase, Type I/genetics , Hep G2 Cells , Mice
8.
Nat Cell Biol ; 26(9): 1470-1481, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39009641

ABSTRACT

Therapy resistance and metastasis, the most fatal steps in cancer, are often triggered by a (partial) activation of the epithelial-mesenchymal transition (EMT) programme. A mesenchymal phenotype predisposes to ferroptosis, a cell death pathway exerted by an iron and oxygen-radical-mediated peroxidation of phospholipids containing polyunsaturated fatty acids. We here show that various forms of EMT activation, including TGFß stimulation and acquired therapy resistance, increase ferroptosis susceptibility in cancer cells, which depends on the EMT transcription factor Zeb1. We demonstrate that Zeb1 increases the ratio of phospholipids containing pro-ferroptotic polyunsaturated fatty acids over cyto-protective monounsaturated fatty acids by modulating the differential expression of the underlying crucial enzymes stearoyl-Co-A desaturase 1 (SCD), fatty acid synthase (FASN), fatty acid desaturase 2 (FADS2), elongation of very long-chain fatty acid 5 (ELOVL5) and long-chain acyl-CoA synthetase 4 (ACSL4). Pharmacological inhibition of selected lipogenic enzymes (SCD and FADS2) allows the manipulation of ferroptosis sensitivity preferentially in high-Zeb1-expressing cancer cells. Our data are of potential translational relevance and suggest a combination of ferroptosis activators and SCD inhibitors for the treatment of aggressive cancers expressing high Zeb1.


Subject(s)
Epithelial-Mesenchymal Transition , Ferroptosis , Phospholipids , Stearoyl-CoA Desaturase , Zinc Finger E-box-Binding Homeobox 1 , Zinc Finger E-box-Binding Homeobox 1/metabolism , Zinc Finger E-box-Binding Homeobox 1/genetics , Humans , Cell Line, Tumor , Phospholipids/metabolism , Stearoyl-CoA Desaturase/metabolism , Stearoyl-CoA Desaturase/genetics , Lipogenesis , Gene Expression Regulation, Neoplastic , Fatty Acid Desaturases/metabolism , Fatty Acid Desaturases/genetics , Animals , Neoplasms/pathology , Neoplasms/metabolism , Neoplasms/genetics , Coenzyme A Ligases/metabolism , Coenzyme A Ligases/genetics , Transforming Growth Factor beta/metabolism , Delta-5 Fatty Acid Desaturase , Drug Resistance, Neoplasm , Fatty Acid Synthase, Type I/metabolism , Fatty Acid Synthase, Type I/genetics
9.
Cell Rep ; 43(8): 114516, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39024103

ABSTRACT

Despite its significance, the role of lipid metabolism in NLRP3 inflammasome remains elusive. Here, we reveal a critical role for fatty acid synthase (FASN) in NLRP3 inflammasome activation. We demonstrate that pharmacological or genetic depletion of FASN dampens NLRP3 activation in primary mouse and human macrophages and in mice. This disruption in NLRP3 activation is contingent upon FASN activity. Accordingly, abolishing cellular palmitoylation, a post-translational modification in which the FASN product palmitate is reversibly conjugated to cysteine residues of target proteins, blunts inflammasome signaling. Correspondingly, an acyl-biotin exchange assay corroborated NLRP3 palmitoylation. Mechanistically, Toll-like receptor (TLR) ligation introduces palmitoylation at NLRP3 Cys898, permitting NLRP3 translocation to dispersed trans-Golgi network (dTGN) vesicles, the site of inflammasome assembly, upon NLRP3 activation. Accordingly, the NLRP3 Cys898 mutant exhibits reduced palmitoylation, limited translocation to the dTGN compartment, and diminished inflammasome activation. These results underscore mechanistic insights through which lipid metabolism licenses NLRP3 inflammasome assembly and activation.


Subject(s)
Inflammasomes , Lipoylation , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , Humans , Mice , Fatty Acid Synthase, Type I/metabolism , Fatty Acid Synthase, Type I/genetics , Fatty Acids/metabolism , Inflammasomes/metabolism , Macrophages/metabolism , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein Transport/drug effects , trans-Golgi Network/metabolism
10.
J Transl Med ; 22(1): 676, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39044184

ABSTRACT

BACKGROUND: Breast cancer manifests as a heterogeneous pathology marked by complex metabolic reprogramming essential to satisfy its energy demands. Oncogenic signals boost the metabolism, modifying fatty acid synthesis and glucose use from the onset to progression and therapy resistant-forms. However, the exact contribution of metabolic dependencies during tumor evolution remains unclear. METHODS: In this study, we elucidate the connection between FASN and LDHA, pivotal metabolic genes, and their correlation with tumor grade and therapy response using datasets from public repositories. Subsequently, we evaluated the metabolic and proliferative functions upon FASN and LDHA inhibition in breast cancer models. Lastly, we integrated metabolomic and lipidomic analysis to define the contributions of metabolites, lipids, and precursors to the metabolic phenotypes. RESULTS: Collectively, our findings indicate metabolic shifts during breast cancer progression, unvealling two distinct functional energy phenotypes associated with aggressiveness and therapy response. Specifically, FASN exhibits reduced expression in advance-grade tumors and therapy-resistant forms, whereas LDHA demonstrates higher expression. Additionally, the biological and metabolic impact of blocking the enzymatic activity of FASN and LDHA was correlated with resistant conditions. CONCLUSIONS: These observations emphasize the intrinsic metabolic heterogeneity within breast cancer, thereby highlighting the relevance of metabolic interventions in the field of precision medicine.


Subject(s)
Breast Neoplasms , Fatty Acid Synthase, Type I , Humans , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/enzymology , Female , Fatty Acid Synthase, Type I/metabolism , Fatty Acid Synthase, Type I/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm , Cell Proliferation , Gene Expression Regulation, Neoplastic , Lipidomics , Metabolomics , L-Lactate Dehydrogenase
11.
Pathol Res Pract ; 260: 155465, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39018927

ABSTRACT

Fatty acid synthase (FASN) is a critical enzyme essential for the production of fats in the body. The abnormal expression of FASN is associated with different types of malignancies, including ovarian cancer. FASN plays a crucial role in cell growth and survival as a metabolic oncogene, although the specific processes that cause its dysregulation are still unknown. FASN interacts with signaling pathways linked to the progression of cancer. Pharmacologically inhibiting or inactivating the FASN gene has shown potential in causing the death of cancer cells, offering a possible treatment approach. This review examines the function of FASN in ovarian cancer, namely its level of expression, influence on the advancement of the disease, and its potential as a target for therapeutic interventions.


Subject(s)
Fatty Acid Synthases , Ovarian Neoplasms , Humans , Ovarian Neoplasms/pathology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/enzymology , Female , Fatty Acid Synthases/metabolism , Fatty Acid Synthases/antagonists & inhibitors , Neoplasm Metastasis , Signal Transduction , Animals , Molecular Targeted Therapy , Fatty Acid Synthase, Type I
12.
Medicina (Kaunas) ; 60(7)2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39064589

ABSTRACT

Background and Objectives: Aberrant upregulation of fatty acid synthase (FASN), catalyzing de novo synthesis of fatty acids, occurs in various tumor types, including human hepatocellular carcinoma (HCC). Although FASN oncogenic activity seems to reside in its pro-lipogenic function, cumulating evidence suggests that FASN's tumor-supporting role might also be metabolic-independent. Materials and Methods: In the present study, we show that FASN inactivation by specific small interfering RNA (siRNA) promoted the downregulation of the S-phase kinase associated-protein kinase 2 (SKP2) and the consequent induction of p27KIP1 in HCC cell lines. Results: Expression levels of FASN and SKP2 directly correlated in human HCC specimens and predicted a dismal outcome. In addition, forced overexpression of SKP2 rendered HCC cells resistant to the treatment with the FASN inhibitor C75. Furthermore, FASN deletion was paralleled by SKP2 downregulation and p27KIP1 induction in the AKT-driven HCC preclinical mouse model. Moreover, forced overexpression of an SKP2 dominant negative form or a p27KIP1 non-phosphorylatable (p27KIP1-T187A) construct completely abolished AKT-dependent hepatocarcinogenesis in vitro and in vivo. Conclusions: In conclusion, the present data indicate that SKP2 is a critical downstream effector of FASN and AKT-dependent hepatocarcinogenesis in liver cancer, envisaging the possibility of effectively targeting FASN-positive liver tumors with SKP2 inhibitors or p27KIP1 activators.


Subject(s)
Carcinoma, Hepatocellular , Cyclin-Dependent Kinase Inhibitor p27 , Liver Neoplasms , S-Phase Kinase-Associated Proteins , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , S-Phase Kinase-Associated Proteins/metabolism , S-Phase Kinase-Associated Proteins/genetics , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Humans , Animals , Mice , Cell Line, Tumor , Fatty Acid Synthases/metabolism , Fatty Acid Synthase, Type I/metabolism , Fatty Acid Synthase, Type I/genetics , Down-Regulation , Male
13.
J Agric Food Chem ; 72(26): 14620-14629, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38885170

ABSTRACT

Milk fat content is a critical indicator of milk quality. Exploring the key regulatory genes involved in milk fat synthesis is essential for enhancing milk fat content. STF-62247 (STF), a thiazolamide compound, has the potential to bind with ALG5 and upregulate lipid droplets in fat synthesis. However, the effect of STF on the process of milk fat synthesis and whether it acts through ALG5 remains unknown. In this study, the impact of ALG5 on milk fat synthesis and its underlying mechanism were investigated using bovine mammary epithelial cells (BMECs) and mouse models through real-time PCR, western blotting, Oil Red O staining, and triglyceride analysis. Experimental findings revealed a positive correlation between STF and ALG5 with the ability to synthesize milk fat. Silencing ALG5 led to decreased expression of FASN, SREBP1, and PPARγ in BMECs, as well as reduced phosphorylation levels in the PI3K/AKT/mTOR signaling pathway. Moreover, the phosphorylation levels of the PI3K/AKT/mTOR signaling pathway were restored when ALG5 silencing was followed by the addition of STF. These results suggest that STF regulates fatty acid synthesis in BMECs by affecting the PI3K/AKT/mTOR signaling pathway through ALG5. ALG5 is possibly a new factor in milk fat synthesis.


Subject(s)
Epithelial Cells , Mammary Glands, Animal , Milk , Signal Transduction , Sterol Regulatory Element Binding Protein 1 , TOR Serine-Threonine Kinases , Animals , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/genetics , Milk/chemistry , Milk/metabolism , Mice , Cattle , Female , Epithelial Cells/metabolism , Mammary Glands, Animal/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Fats/metabolism , PPAR gamma/metabolism , PPAR gamma/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Fatty Acids/metabolism , Fatty Acid Synthase, Type I/genetics , Fatty Acid Synthase, Type I/metabolism , Triglycerides/metabolism
14.
Cell Biochem Biophys ; 82(3): 2285-2296, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38824236

ABSTRACT

Fatty acid synthase (FASN) catalyzes the rate-limiting step of cellular lipogenesis. FASN expression is upregulated in various types of cancer cells, implying that FASN is a potential target for cancer therapy. 2-Deoxy-D-glucose (2-DG) specifically targets cancer cells by inhibiting glycolysis and glucose metabolism, resulting in multiple anticancer effects. However, whether the effects of 2-DG involve lipogenic metabolism remains to be elucidated. We investigated the effect of 2-DG administration on FASN expression in HeLa human cervical cancer cells. 2-DG treatment for 24 h decreased FASN mRNA and protein levels and suppressed the activity of an exogenous rat Fasn promoter. The use of a chemical activator or inhibitors or of a mammalian expression plasmid showed that neither AMPK nor the Sp1 transcription factor is responsible for the inhibitory effect of 2-DG on FASN expression. Administration of thapsigargin, an endoplasmic reticulum (ER) stress inducer, or 4-(2-aminoethyl) benzenesulfonyl fluoride (AEBSF), a site 1 protease inhibitor, mimicked the inhibitory effect of 2-DG on FASN expression. 2-DG did not further decrease FASN expression in the presence of thapsigargin or AEBSF. Site 1 protease mediates activation of ATF6, an ER stress mediator, as well as sterol regulatory element-binding protein 1 (SREBP1), a robust transcription factor for FASN. Administration of 2-DG or thapsigargin for 24 h suppressed activation of ATF6 and SREBP1, as did AEBSF. We speculated that these effects of 2-DG or thapsigargin are due to feedback inhibition via increased GRP78 expression following ER stress. Supporting this, exogenous overexpression of GRP78 in HeLa cells suppressed SREBP1 activation and Fasn promoter activity. These results suggest that 2-DG suppresses FASN expression via an ER stress-dependent pathway, providing new insight into the molecular basis of FASN regulation in cancer.


Subject(s)
Deoxyglucose , Down-Regulation , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Animals , Humans , Rats , Activating Transcription Factor 6/metabolism , Activating Transcription Factor 6/genetics , AMP-Activated Protein Kinases/metabolism , Deoxyglucose/pharmacology , Down-Regulation/drug effects , Endoplasmic Reticulum Stress/drug effects , Fatty Acid Synthase, Type I/metabolism , Fatty Acid Synthase, Type I/genetics , Fatty Acid Synthases/metabolism , Fatty Acid Synthases/genetics , HeLa Cells , Promoter Regions, Genetic , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Thapsigargin/pharmacology
15.
Biochim Biophys Acta Mol Basis Dis ; 1870(7): 167299, 2024 10.
Article in English | MEDLINE | ID: mdl-38878833

ABSTRACT

STING (stimulator of interferon genes) is a critical immunoregulatory protein in sepsis and is regulated by various mechanisms, especially palmitoylation. FASN (fatty acid synthase) is the rate-limiting enzyme to generate cellular palmitic acid (PA) via acetyl-CoA and malonyl-CoA and participates in protein palmitoylation. However, the mechanisms underlying the interaction between STING and FASN have not been completely understood. In this study, STING-knockout mice were used to confirm the pivotal role of STING in sepsis-induced liver injury. Metabolomics confirmed the dyslipidemia in septic mice and patients. The compounds library was screened, revealing that FASN inhibitors exerted a significant inhibitory effect on the STING pathway. Mechanically, the regulatory effect of FASN on the STING pathway was dependent on palmitoylation. Further experiments indicated that the upstream of FASN, malonyl-CoA inhibited STING pathway possibly due to C91 (palmitoylated residue) of STING. Overall, this study reveals a novel paradigm of STING regulation and provides a new perspective on immunity and metabolism.


Subject(s)
Fatty Acid Synthase, Type I , Lipoylation , Macrophages , Malonyl Coenzyme A , Membrane Proteins , Sepsis , Animals , Humans , Male , Mice , Fatty Acid Synthase, Type I/metabolism , Fatty Acid Synthase, Type I/genetics , Liver/metabolism , Liver/pathology , Macrophages/metabolism , Malonyl Coenzyme A/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Palmitic Acid/pharmacology , Sepsis/metabolism , Sepsis/complications , Sepsis/drug therapy , Signal Transduction/drug effects
16.
Int J Mol Sci ; 25(9)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38732103

ABSTRACT

Fatty acid synthesis has been extensively investigated as a therapeutic target in cancers, including colorectal cancer (CRC). Fatty acid synthase (FASN), a key enzyme of de novo lipid synthesis, is significantly upregulated in CRC, and therapeutic approaches of targeting this enzyme are currently being tested in multiple clinical trials. However, the mechanisms behind the pro-oncogenic action of FASN are still not completely understood. Here, for the first time, we show that overexpression of FASN increases the expression of glutamine-fructose-6-phosphate transaminase 1 (GFPT1) and O-linked N-acetylglucosamine transferase (OGT), enzymes involved in hexosamine metabolism, and the level of O-GlcNAcylation in vitro and in vivo. Consistently, expression of FASN significantly correlates with expression of GFPT1 and OGT in human CRC tissues. shRNA-mediated downregulation of GFPT1 and OGT inhibits cellular proliferation and the level of protein O-GlcNAcylation in vitro, and knockdown of GFPT1 leads to a significant decrease in tumor growth and metastasis in vivo. Pharmacological inhibition of GFPT1 and OGT leads to significant inhibition of cellular proliferation and colony formation in CRC cells. In summary, our results show that overexpression of FASN increases the expression of GFPT1 and OGT as well as the level of protein O-GlcNAcylation to promote progression of CRC; targeting the hexosamine biosynthesis pathway could be a therapeutic approach for this disease.


Subject(s)
Cell Proliferation , Colorectal Neoplasms , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing) , N-Acetylglucosaminyltransferases , Humans , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/genetics , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/metabolism , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/genetics , N-Acetylglucosaminyltransferases/metabolism , N-Acetylglucosaminyltransferases/genetics , Glycosylation , Animals , Mice , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Up-Regulation , Mice, Nude , Fatty Acid Synthase, Type I
17.
Cell Signal ; 120: 111232, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38763183

ABSTRACT

Aging affects lipid metabolism and can cause obesity as it is closely related to the disorder of many lipogenic regulatory factors. LncRNAs have been recognized as pivotal regulators across diverse biological processes, but their effects on lipogenesis in aging remain to be further studied. In this work, using RNA sequencing (RNA-Seq), we found that the expression of lncRNA AI504432 was significantly upregulated in the eWAT (epididymal white adipose tissue) of aging mice, and the knockdown of AI504432 notably reduced the expression of several adipogenic genes (e.g., Cebp/α, Srebp-1c, Fasn, Acaca, and Scd1) in senescent adipocytes. The bioinformatics investigation revealed that AI504432 possessed a binding site for miR-1a-3p, and the discovery was verified by the luciferase reporter assay. The expression of Fasn was increased upon the inhibition of miR-1a-3p but restored upon the simultaneous silencing of AI504432. Taken together, our results suggested that AI504432 controlled lipogenesis through the miR-1a-3p/Fasn signaling pathway. The findings may inspire new therapeutic approaches to target imbalanced lipid homeostasis due to aging.


Subject(s)
Adipocytes , Cellular Senescence , Fatty Acid Synthase, Type I , Lipogenesis , MicroRNAs , RNA, Long Noncoding , Up-Regulation , Animals , MicroRNAs/metabolism , MicroRNAs/genetics , Lipogenesis/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Mice , Adipocytes/metabolism , Fatty Acid Synthase, Type I/metabolism , Fatty Acid Synthase, Type I/genetics , Up-Regulation/drug effects , Male , Mice, Inbred C57BL , Aging/metabolism , Aging/genetics
18.
Sci Adv ; 10(20): eadj5942, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38758779

ABSTRACT

Acetyl-CoA synthetase short-chain family member 1 (ACSS1) uses acetate to generate mitochondrial acetyl-CoA and is regulated by deacetylation by sirtuin 3. We generated an ACSS1-acetylation (Ac) mimic mouse, where lysine-635 was mutated to glutamine (K635Q). Male Acss1K635Q/K635Q mice were smaller with higher metabolic rate and blood acetate and decreased liver/serum ATP and lactate levels. After a 48-hour fast, Acss1K635Q/K635Q mice presented hypothermia and liver aberrations, including enlargement, discoloration, lipid droplet accumulation, and microsteatosis, consistent with nonalcoholic fatty liver disease (NAFLD). RNA sequencing analysis suggested dysregulation of fatty acid metabolism, cellular senescence, and hepatic steatosis networks, consistent with NAFLD. Fasted Acss1K635Q/K635Q mouse livers showed increased fatty acid synthase (FASN) and stearoyl-CoA desaturase 1 (SCD1), both associated with NAFLD, and increased carbohydrate response element-binding protein binding to Fasn and Scd1 enhancer regions. Last, liver lipidomics showed elevated ceramide, lysophosphatidylethanolamine, and lysophosphatidylcholine, all associated with NAFLD. Thus, we propose that ACSS1-K635-Ac dysregulation leads to aberrant lipid metabolism, cellular senescence, and NAFLD.


Subject(s)
Acetate-CoA Ligase , Cellular Senescence , Mitochondria , Non-alcoholic Fatty Liver Disease , Stearoyl-CoA Desaturase , Animals , Male , Mice , Acetate-CoA Ligase/metabolism , Acetate-CoA Ligase/genetics , Acetylation , Cellular Senescence/genetics , Coenzyme A Ligases , Disease Models, Animal , Fatty Acid Synthase, Type I , Gene Knock-In Techniques , Lipid Metabolism , Liver/metabolism , Liver/pathology , Mitochondria/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Sirtuin 3/metabolism , Sirtuin 3/genetics , Stearoyl-CoA Desaturase/metabolism , Stearoyl-CoA Desaturase/genetics
19.
J Agric Food Chem ; 72(23): 13069-13082, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38809951

ABSTRACT

Intestinal fibrosis is a common complication of Crohn's disease and characterized by excessive extracellular matrix (ECM) deposition. The aryl hydrocarbon receptor (AhR) detects micronutrients and microbial metabolites in diet and can attenuate intestinal fibrosis with unclear mechanisms. In this study, AhR activation was demonstrated to downregulate the transcription of collagen I and fibronectin in a Sp1- but not Sp3- or AP-1-dependent manner. A suppressed fatty acid synthesis was highlighted using untargeted metabolomics analyses, and synthetic products, palmitic acid (PA), were used as the intermediary agent. After a screening study, fatty acid synthase (FASN) was identified as the main targeted protein, and AhR activation regulated "HDAC3-acetylation" signals but not glycosylation to enhance FASN degradation. Furthermore, results of bioinformatics analysis and others showed that after being activated, AhR targeted miR-193a-3p to control HDAC3 transcription. Collectively, AhR activation inhibited ECM deposition and alleviated intestinal fibrosis by limiting fatty acid synthesis subsequent to the inhibition of "miR-193a-3p-HDAC3-FASN" signals.


Subject(s)
Fatty Acids , Fibrosis , Histone Deacetylases , Intestines , Receptors, Aryl Hydrocarbon , Animals , Humans , Male , Mice , Fatty Acid Synthase, Type I/metabolism , Fatty Acid Synthase, Type I/genetics , Fatty Acids/metabolism , Fibrosis/metabolism , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Intestinal Mucosa/metabolism , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Aryl Hydrocarbon/genetics , Signal Transduction
20.
Biochim Biophys Acta Mol Basis Dis ; 1870(6): 167258, 2024 08.
Article in English | MEDLINE | ID: mdl-38788910

ABSTRACT

The increasing prevalence of obesity, type 2 diabetes mellitus (T2DM), and gestational diabetes (GDM) among pregnant women has risen dramatically worldwide. The antihyperglycemic drug metformin is the most common drug for T2DM treatment in non-pregnant individuals; nevertheless, it is increasingly being used for diabetes-complicated pregnancies. Studies on the long-term metabolic effects of this drug in offspring remain scarce. This work aimed to determine the effect of metformin exposure during pregnancy and lactation on the offspring of a model of diet-induced maternal hyperglycemia. Cohorts of pregnant mice were fed a 46% fat diet (HFD) or a control standard diet (SD). A group of dams were exposed to metformin during pregnancy and lactation. After weaning, the offspring were fed SD for 8 weeks and then challenged with a 46% HFD after puberty for 12 weeks. Irrespective of the maternal diet, offspring of metformin-exposed mothers had a lower body weight and reduced inguinal white adipose tissue (iWAT) mass after HFD challenge. This was associated with increased expression of Pparg, Fabp4, Glut4, Srebp1, and Fasn in the iWAT during adulthood in the metabolically impaired dams exposed to metformin, suggesting increased adipogenesis and de novo lipogenesis. Increased expression of Fasn associated with decreased methylation levels at its promoter and proximal coding region in the iWAT was found. These results suggest that metformin modulates gene expression levels by epigenetic mechanisms in maternal metabolic-impaired conditions.


Subject(s)
Body Weight , Diet, High-Fat , Lactation , Metformin , Prenatal Exposure Delayed Effects , Sterol Regulatory Element Binding Protein 1 , Animals , Metformin/pharmacology , Female , Pregnancy , Lactation/drug effects , Mice , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/pathology , Prenatal Exposure Delayed Effects/chemically induced , Diet, High-Fat/adverse effects , Body Weight/drug effects , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , PPAR gamma/metabolism , PPAR gamma/genetics , Glucose Transporter Type 4/metabolism , Glucose Transporter Type 4/genetics , Hypoglycemic Agents/pharmacology , Adipose Tissue, White/metabolism , Adipose Tissue, White/drug effects , Obesity/metabolism , Obesity/pathology , Obesity/chemically induced , Fatty Acid Synthase, Type I/metabolism , Fatty Acid Synthase, Type I/genetics , Male , Mice, Inbred C57BL , Adipose Tissue/metabolism , Adipose Tissue/drug effects , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL