Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 844
Filter
1.
BMC Immunol ; 25(1): 33, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38834979

ABSTRACT

PURPOSE: Severe community-acquired pneumonia (SCAP) is a common respiratory system disease with rapid development and high mortality. Exploring effective biomarkers for early detection and development prediction of SCAP is of urgent need. The function of miR-486-5p in SCAP diagnosis and prognosis was evaluated to identify a promising biomarker for SCAP. PATIENTS AND METHODS: The serum miR-486-5p in 83 patients with SCAP, 52 healthy individuals, and 68 patients with mild CAP (MCAP) patients were analyzed by PCR. ROC analysis estimated miR-486-5p in screening SCAP, and the Kaplan-Meier and Cox regression analyses evaluated the predictive value of miR-486-5p. The risk factors for MCAP patients developing SCAP were assessed by logistic analysis. The alveolar epithelial cell was treated with Klebsiella pneumonia to mimic the occurrence of SCAP. The targeting mechanism underlying miR-486-5p was evaluated by luciferase reporter assay. RESULTS: Upregulated serum miR-486-5p screened SCAP from healthy individuals and MCAP patients with high sensitivity and specificity. Increasing serum miR-486-5p predicted the poor outcomes of SCAP and served as a risk factor for MCAP developing into SCAP. K. pneumonia induced suppressed proliferation, significant inflammation and oxidative stress in alveolar epithelial cells, and silencing miR-486-5p attenuated it. miR-486-5p negatively regulated FOXO1, and the knockdown of FOXO1 reversed the effect of miR-486-5p in K. pneumonia-treated alveolar epithelial cells. CONCLUSION: miR-486-5p acted as a biomarker for the screening and monitoring of SCAP and predicting the malignancy of MCAP. Silencing miR-486-5p alleviated inflammation and oxidative stress induced by K. pneumonia via negatively modulating FOXO1.


Subject(s)
Community-Acquired Infections , Forkhead Box Protein O1 , Klebsiella Infections , MicroRNAs , Humans , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , MicroRNAs/genetics , Community-Acquired Infections/diagnosis , Male , Female , Middle Aged , Klebsiella Infections/diagnosis , Prognosis , Biomarkers , Klebsiella pneumoniae/physiology , Aged , Risk Factors , Alveolar Epithelial Cells/metabolism , Pneumonia/genetics , Oxidative Stress/genetics
2.
Cells ; 13(9)2024 May 02.
Article in English | MEDLINE | ID: mdl-38727314

ABSTRACT

During the secretory phase of the menstrual cycle, endometrial fibroblast cells begin to change into large epithelial-like cells called decidual cells in a process called decidualization. This differentiation continues more broadly in the endometrium and forms the decidual tissue during early pregnancy. The cells undergoing decidualization as well as the resulting decidual cells, support successful implantation and placentation during early pregnancy. This study was carried out to identify new potentially important long non-coding RNA (lncRNA) genes that may play a role in human endometrial stromal fibroblast cells (hESF) undergoing decidualization in vitro, and several were found. The expression of nine was further characterized. One of these, AC027288.3, showed a dramatic increase in the expression of hESF cells undergoing decidualization. When AC027288.3 expression was targeted, the ability of the cells to undergo decidualization as determined by the expression of decidualization marker protein-coding genes was significantly altered. The most affected markers of decidualization whose expression was significantly reduced were FOXO1, FZD4, and INHBA. Therefore, AC027288.3 may be a major upstream regulator of the WNT-FOXO1 pathway and activin-SMAD3 pathways previously shown as critical for hESF decidualization. Finally, we explored possible regulators of AC027288.3 expression during human ESF decidualization. Expression was regulated by cAMP and progesterone. Our results suggest that AC027288.3 plays a role in hESF decidualization and identifies several other lncRNA genes that may also play a role.


Subject(s)
Decidua , Endometrium , Fibroblasts , RNA, Long Noncoding , Stromal Cells , Humans , Female , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Fibroblasts/metabolism , Fibroblasts/cytology , Decidua/metabolism , Decidua/cytology , Endometrium/cytology , Endometrium/metabolism , Stromal Cells/metabolism , Stromal Cells/cytology , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/genetics , Pregnancy , Adult , Cell Differentiation/genetics
3.
Biol Res ; 57(1): 27, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38745315

ABSTRACT

BACKGROUND: Aberrant gluconeogenesis is considered among primary drivers of hyperglycemia under insulin resistant conditions, with multiple studies pointing towards epigenetic dysregulation. Here we examine the role of miR-721 and effect of epigenetic modulator laccaic acid on the regulation of gluconeogenesis under high fat diet induced insulin resistance. RESULTS: Reanalysis of miRNA profiling data of high-fat diet-induced insulin-resistant mice model, GEO dataset (GSE94799) revealed a significant upregulation of miR-721, which was further validated in invivo insulin resistance in mice and invitro insulin resistance in Hepa 1-6 cells. Interestingly, miR-721 mimic increased glucose production in Hepa 1-6 cells via activation of FOXO1 regulated gluconeogenic program. Concomitantly, inhibition of miR-721 reduced glucose production in palmitate induced insulin resistant Hepa 1-6 cells by blunting the FOXO1 induced gluconeogenesis. Intriguingly, at epigenetic level, enrichment of the transcriptional activation mark H3K36me2 got decreased around the FOXO1 promoter. Additionally, identifying targets of miR-721 using miRDB.org showed H3K36me2 demethylase KDM2A as a potential target. Notably, miR-721 inhibitor enhanced KDM2A expression which correlated with H3K36me2 enrichment around FOXO1 promoter and the downstream activation of the gluconeogenic pathway. Furthermore, inhibition of miR-721 in high-fat diet-induced insulin-resistant mice resulted in restoration of KDM2A levels, concomitantly reducing FOXO1, PCK1, and G6PC expression, attenuating gluconeogenesis, hyperglycemia, and improving glucose tolerance. Interestingly, the epigenetic modulator laccaic acid also reduced the hepatic miR-721 expression and improved KDM2A expression, supporting our earlier report that laccaic acid attenuates insulin resistance by reducing gluconeogenesis. CONCLUSION: Our study unveils the role of miR-721 in regulating gluconeogenesis through KDM2A and FOXO1 under insulin resistance, pointing towards significant clinical and therapeutic implications for metabolic disorders. Moreover, the promising impact of laccaic acid highlights its potential as a valuable intervention in managing insulin resistance-associated metabolic diseases.


Subject(s)
Diet, High-Fat , Epigenesis, Genetic , Gluconeogenesis , Insulin Resistance , Jumonji Domain-Containing Histone Demethylases , Mice, Inbred C57BL , MicroRNAs , Animals , Insulin Resistance/physiology , Gluconeogenesis/genetics , Gluconeogenesis/physiology , MicroRNAs/metabolism , MicroRNAs/genetics , Mice , Jumonji Domain-Containing Histone Demethylases/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Male , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/genetics
5.
Braz J Med Biol Res ; 57: e13378, 2024.
Article in English | MEDLINE | ID: mdl-38716982

ABSTRACT

Forkhead Box O1 (FOXO1) has been reported to play important roles in many tumors. However, FOXO1 has not been studied in pan-cancer. The purpose of this study was to reveal the roles of FOXO1 in pan-cancer (33 cancers in this study). Through multiple public platforms, a pan-cancer analysis of FOXO1 was conducted to obtained FOXO1 expression profiles in various tumors to explore the relationship between FOXO1 expression and prognosis of these tumors and to disclose the potential mechanism of FOXO1 in these tumors. FOXO1 was associated with the prognosis of multiple tumors, especially LGG (low grade glioma), OV (ovarian carcinoma), and KIRC (kidney renal clear cell carcinoma). FOXO1 might play the role of an oncogenic gene in LGG and OV, while playing the role of a cancer suppressor gene in KIRC. FOXO1 expression had a significant correlation with the infiltration of some immune cells in LGG, OV, and KIRC. By combining FOXO1 expression and immune cell infiltration, we found that FOXO1 might influence the overall survival of LGG through the infiltration of myeloid dendritic cells or CD4+ T cells. Functional enrichment analysis and gene set enrichment analysis showed that FOXO1 might play roles in tumors through immunoregulatory interactions between a lymphoid and a non-lymphoid cell, TGF-beta signaling pathway, and transcriptional misregulation in cancer. FOXO1 was associated with the prognosis of multiple tumors, especially LGG, OV, and KIRC. In these tumors, FOXO1 might play its role via the regulation of the immune microenvironment.


Subject(s)
Forkhead Box Protein O1 , Neoplasms , Humans , Biomarkers, Tumor/genetics , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Gene Expression Regulation, Neoplastic , Neoplasms/immunology , Neoplasms/genetics , Prognosis , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics
6.
Sci Rep ; 14(1): 11497, 2024 05 20.
Article in English | MEDLINE | ID: mdl-38769106

ABSTRACT

Barth syndrome (BTHS) is a rare disorder caused by mutations in the TAFAZZIN gene. Previous studies from both patients and model systems have established metabolic dysregulation as a core component of BTHS pathology. In particular, features such as lactic acidosis, pyruvate dehydrogenase (PDH) deficiency, and aberrant fatty acid and glucose oxidation have been identified. However, the lack of a mechanistic understanding of what causes these conditions in the context of BTHS remains a significant knowledge gap, and this has hindered the development of effective therapeutic strategies for treating the associated metabolic problems. In the current study, we utilized tafazzin-knockout C2C12 mouse myoblasts (TAZ-KO) and cardiac and skeletal muscle tissue from tafazzin-knockout mice to identify an upstream mechanism underlying impaired PDH activity in BTHS. This mechanism centers around robust upregulation of pyruvate dehydrogenase kinase 4 (PDK4), resulting from hyperactivation of AMP-activated protein kinase (AMPK) and subsequent transcriptional upregulation by forkhead box protein O1 (FOXO1). Upregulation of PDK4 in tafazzin-deficient cells causes direct phospho-inhibition of PDH activity accompanied by increased glucose uptake and elevated intracellular glucose concentration. Collectively, our findings provide a novel mechanistic framework whereby impaired tafazzin function ultimately results in robust PDK4 upregulation, leading to impaired PDH activity and likely linked to dysregulated metabolic substrate utilization. This mechanism may underlie previously reported findings of BTHS-associated metabolic dysregulation.


Subject(s)
AMP-Activated Protein Kinases , Forkhead Box Protein O1 , Mice, Knockout , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Animals , Mice , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/genetics , AMP-Activated Protein Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Up-Regulation , Signal Transduction , Myoblasts/metabolism , Cell Line , Glucose/metabolism , Acyltransferases
7.
Cell Mol Life Sci ; 81(1): 237, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38795132

ABSTRACT

Ovarian endometriosis is a common gynecological disease, and one of its most significant symptoms is infertility. In patients with endometriosis, defects in endometrial decidualization lead to impaired endometrial receptivity and embryo implantation, thus affecting early pregnancy and women's desire to have children. However, the mechanisms underlying the development of endometriosis and its associated defective decidualization are unclear. We find that NEK2 expression is increased in the ectopic and eutopic endometrium of patients with endometriosis. Meanwhile, NEK2 interacts with FOXO1 and phosphorylates FOXO1 at Ser184, inhibiting the stability of the FOXO1 protein. Importantly, NEK2-mediated phosphorylation of FOXO1 at Ser184 promotes cell proliferation, migration, invasion and impairs decidualization. Furthermore, INH1, an inhibitor of NEK2, inhibits the growth of ectopic lesions in mouse models of endometriosis and promotes endometrial decidualization in mouse models of artificially induced decidualization. Taken together, these findings indicate that NEK2 regulates the development of endometriosis and associated disorders of decidualization through the phosphorylation of FOXO1, providing a new therapeutic target for its treatment.


Subject(s)
Cell Proliferation , Endometriosis , Endometrium , Forkhead Box Protein O1 , NIMA-Related Kinases , Female , Endometriosis/metabolism , Endometriosis/pathology , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/genetics , Humans , Animals , Phosphorylation , Mice , NIMA-Related Kinases/metabolism , NIMA-Related Kinases/genetics , Endometrium/metabolism , Endometrium/pathology , Cell Movement , Decidua/metabolism , Decidua/pathology , Adult , Disease Models, Animal
8.
Sci Rep ; 14(1): 11688, 2024 05 22.
Article in English | MEDLINE | ID: mdl-38778150

ABSTRACT

Prostate cancer lineage plasticity is a key driver in the transition to neuroendocrine prostate cancer (NEPC), and the RTK/RAS signaling pathway is a well-established cancer pathway. Nevertheless, the comprehensive link between the RTK/RAS signaling pathway and lineage plasticity has received limited investigation. In particular, the intricate regulatory network governing the interplay between RTK/RAS and lineage plasticity remains largely unexplored. The multi-omics data were clustered with the coefficient of argument and neighbor joining algorithm. Subsequently, the clustered results were analyzed utilizing the GSEA, gene sets related to stemness, multi-lineage state datasets, and canonical cancer pathway gene sets. Finally, a comprehensive exploration of the data based on the ssGSEA, WGCNA, GSEA, VIPER, prostate cancer scRNA-seq data, and the GPSAdb database was conducted. Among the six modules in the clustering results, there are 300 overlapping genes, including 3 previously unreported prostate cancer genes that were validated to be upregulated in prostate cancer through RT-qPCR. Function Module 6 shows a positive correlation with prostate cancer cell stemness, multi-lineage states, and the RTK/RAS signaling pathway. Additionally, the 19 leading-edge genes of the RTK/RAS signaling pathway promote prostate cancer lineage plasticity through a complex network of transcriptional regulation and copy number variations. In the transcriptional regulation network, TP63 and FOXO1 act as suppressors of prostate cancer lineage plasticity, whereas RORC exerts a promoting effect. This study provides a comprehensive perspective on the role of the RTK/RAS pathway in prostate cancer lineage plasticity and offers new clues for the treatment of NEPC.


Subject(s)
Data Mining , Prostatic Neoplasms , Signal Transduction , Transcription Factors , Male , Humans , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , Signal Transduction/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , ras Proteins/genetics , ras Proteins/metabolism , DNA Copy Number Variations , Gene Expression Regulation, Neoplastic , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Gene Regulatory Networks , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Cell Lineage/genetics
9.
Arch Dermatol Res ; 316(6): 274, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38796528

ABSTRACT

Wound healing is a highly programmed process, in which any abnormalities result in scar formation. MicroRNAs are potent regulators affecting wound repair and scarification. However, the function of microRNAs in wound healing is not fully understood. Here, we analyzed the expression and function of microRNAs in patients with cutaneous wounds. Cutaneous wound biopsies from patients with either hypertrophic scarring or normal wound repair were collected during inflammation, proliferation, and remodeling phases. Fourteen candidate microRNAs were selected for expression analysis by qRT-PCR. The expression of genes involved in inflammation, angiogenesis, proliferation, and migration were measured using qRT-PCR. Cell cycle and scratch assays were used to explore the proliferation and migration rates. Flow cytometry analysis was employed to examine TGF-ß, αSMA and collagen-I expression. Target gene suggestion was performed using Enrichr tool. The results showed that miR-16-5p, miR-152-3p, miR-125b-5p, miR-34c-5p, and miR-182-5p were revealed to be differentially expressed between scarring and non-scarring wounds. Based on the expression patterns obtained, miR-182-5p was selected for functional studies. miR-182-5p induced RELA expression synergistically upon IL-6 induction in keratinocytes and promoted angiogenesis. miR-182-5p prevented keratinocyte migration, while overexpressed TGF-ß3 following induction of inflammation. Moreover, miR-182-5p enhanced fibroblast proliferation, migration, differentiation, and collagen-1 expression. FoxO1 and FoxO3 were found to potentially serve as putative gene targets of miR-182-5p. In conclusion, miR-182-5p is differentially expressed between scarring and non-scarring wounds and affect the behavior of cells involved in cutaneous wound healing. Deregulated expression of miR-182-5p adversely affects the proper transition of wound healing phases, resulting in scar formation.


Subject(s)
Cell Proliferation , Cicatrix, Hypertrophic , MicroRNAs , Skin , Wound Healing , MicroRNAs/genetics , MicroRNAs/metabolism , Humans , Wound Healing/genetics , Cell Proliferation/genetics , Skin/pathology , Skin/injuries , Skin/metabolism , Cicatrix, Hypertrophic/genetics , Cicatrix, Hypertrophic/pathology , Cicatrix, Hypertrophic/metabolism , Cell Movement/genetics , Inflammation/genetics , Inflammation/pathology , Keratinocytes/metabolism , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/genetics , Male , Female , Adult , Transcription Factor RelA/metabolism , Transcription Factor RelA/genetics , Fibroblasts/metabolism , Gene Expression Regulation , Middle Aged , Neovascularization, Physiologic/genetics
10.
J Agric Food Chem ; 72(22): 12582-12595, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38788215

ABSTRACT

Renal tubular ectopic lipid deposition (ELD) plays a significant role in the development of chronic kidney disease, posing a great threat to human health. The present work aimed to explore the intervention effect and potential molecular mechanism of a purified tea polysaccharide (TPS3A) on renal tubular ELD. The results demonstrated that TPS3A effectively improved kidney function and slowed the progression of tubulointerstitial fibrosis in high-fat-diet (HFD)-exposed ApoE-/- mice. Additionally, TPS3A notably suppressed lipogenesis and enhanced lipolysis, as shown by the downregulation of lipogenesis markers (SREBP-1 and FAS) and the upregulation of lipolysis markers (HSL and ATGL), thereby reducing renal tubular ELD in HFD-fed ApoE-/- mice and palmitic-acid-stimulated HK-2 cells. The AMPK-SIRT1-FoxO1 axis is a core signal pathway in regulating lipid deposition. Consistently, TPS3A significantly increased the levels of phosphorylated-AMPK, SIRT1, and deacetylation of Ac-FoxO1. However, these effects of TPS3A on lipogenesis and lipolysis were abolished by AMPK siRNA, SIRT1 siRNA, and FoxO1 inhibitor, resulting in exacerbated lipid deposition. Taken together, TPS3A shows promise in ameliorating renal tubular ELD by inhibiting lipogenesis and promoting lipolysis through the AMPK-SIRT1-FoxO1 signaling pathway.


Subject(s)
Diet, High-Fat , Lipogenesis , Lipolysis , Mice, Inbred C57BL , Polysaccharides , Animals , Lipogenesis/drug effects , Mice , Lipolysis/drug effects , Male , Diet, High-Fat/adverse effects , Humans , Polysaccharides/pharmacology , Polysaccharides/administration & dosage , Sirtuin 1/metabolism , Sirtuin 1/genetics , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/genetics , Kidney Tubules/metabolism , Kidney Tubules/drug effects , Camellia sinensis/chemistry , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/genetics , Plant Extracts/pharmacology , Plant Extracts/administration & dosage , Tea/chemistry , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 1/genetics
11.
J Clin Invest ; 134(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38747287

ABSTRACT

Lymphedema is a debilitating disease with no effective cure and affects an estimated 250 million individuals worldwide. Prior studies have identified mutations in piezo-type mechanosensitive ion channel component 1 (PIEZO1), angiopoietin 2 (ANGPT2), and tyrosine kinase with Ig-like and EGF-like domains 1 (TIE1) in patients with primary lymphedema. Here, we identified crosstalk between these molecules and showed that activation of the mechanosensory channel PIEZO1 in lymphatic endothelial cells (LECs) caused rapid exocytosis of the TIE ligand ANGPT2, ectodomain shedding of TIE1 by disintegrin and metalloproteinase domain-containing protein 17 (ADAM17), and increased TIE/PI3K/AKT signaling, followed by nuclear export of the transcription factor FOXO1. These data establish a functional network between lymphedema-associated genes and provide what we believe to be the first molecular mechanism bridging channel function with vascular signaling and intracellular events culminating in transcriptional regulation of genes expressed in LECs. Our study provides insights into the regulation of lymphatic function and molecular pathways involved in human disease.


Subject(s)
Angiopoietin-2 , Forkhead Box Protein O1 , Ion Channels , Lymphangiogenesis , Lymphedema , Receptor, TIE-1 , Signal Transduction , Animals , Humans , Mice , ADAM17 Protein/metabolism , ADAM17 Protein/genetics , Angiopoietin-2/metabolism , Angiopoietin-2/genetics , Endothelial Cells/metabolism , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/genetics , Ion Channels/metabolism , Ion Channels/genetics , Lymphangiogenesis/genetics , Lymphedema/metabolism , Lymphedema/genetics , Lymphedema/pathology , Mechanotransduction, Cellular , Receptor, TIE-1/metabolism , Receptor, TIE-1/genetics
12.
Cell Genom ; 4(4): 100537, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38604128

ABSTRACT

Transcriptional dysregulation is a hallmark of diffuse large B cell lymphoma (DLBCL), as transcriptional regulators are frequently mutated. However, our mechanistic understanding of how normal transcriptional programs are co-opted in DLBCL has been hindered by a lack of methodologies that provide the temporal resolution required to separate direct and indirect effects on transcriptional control. We applied a chemical-genetic approach to engineer the inducible degradation of the transcription factor FOXO1, which is recurrently mutated (mFOXO1) in DLBCL. The combination of rapid degradation of mFOXO1, nascent transcript detection, and assessment of chromatin accessibility allowed us to identify the direct targets of mFOXO1. mFOXO1 was required to maintain accessibility at specific enhancers associated with multiple oncogenes, and mFOXO1 degradation impaired RNA polymerase pause-release at some targets. Wild-type FOXO1 appeared to weakly regulate many of the same targets as mFOXO1 and was able to complement the degradation of mFOXO1 in the context of AKT inhibition.


Subject(s)
Forkhead Box Protein O1 , Regulatory Sequences, Nucleic Acid , Humans , Forkhead Box Protein O1/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Transcription Factors/genetics
13.
Discov Med ; 36(183): 699-713, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38665019

ABSTRACT

BACKGROUND: The usage of life-saving mechanical ventilation (MV) could cause ventilator-induced diaphragmatic dysfunction (VIDD), increasing both mortality and morbidity. Aminophylline (AP) has the potential to enhance the contractility of animal skeletal muscle fibers and improve the activity of human respiratory muscles, and the insulin-like growth factor-1 (IGF-1)- forkhead box protein O1 (FOXO1)-muscle RING finger-1 (MURF1) pathway plays a crucial role in skeletal muscle dysfunction. This study aimed to investigate the impact of AP on VIDD and to elucidate the role of the IGF-1-FOXO1-MURF1 pathway as an underlying mechanism. METHODS: Rat models of VIDD were established through MV treatment. IGF-1 lentiviral (LV) interference (LV-IGF-1-shRNA; controlled by lentiviral negative control LV-NC) was employed to inhibit IGF-1 expression and thereby block the IGF-1-FOXO1-MURF1 pathway. Protein and mRNA levels of IGF-1, FOXO1, and MURF1 were assessed using western blot and real-time reverse transcriptase-polymerase chain reaction (RT-qPCR), respectively. Diaphragm contractility and morphometry were examined through measurement of compound muscle action potentials (CMAPs) and hematoxylin and eosin (H&E) staining. Oxidative stress was evaluated by levels of hydrogen peroxide (H2O2), superoxide dismutase (SOD), antioxidant glutathione (GSH), and carbonylated protein. Mitochondrial stability was assessed by measuring the mitochondrial membrane potential (MMP), and mitochondrial fission and mitophagy were examined through protein levels of dynamin-related protein 1 (DRP1), mitofusin 2 protein (MFN2), phosphatase and tensin homolog (PTEN)-induced kinase 1 (PINK1), and Parkin (western blot). Apoptosis was evaluated using the terminal deoxynucleotidyl transferase-mediated uridine 5'-triphosphate (UTP) nick-end labeling (TUNEL) assay and levels of Bax, B-cell lymphoma 2 (BCL-2), and Caspase-3. Levels of Atrogin-1, neuronally expressed developmentally downregulated 4 (NEDD4), and muscle ubiquitin ligase of SCF complex in atrophy-1 (MUSA1) mRNA, as well as ubiquitinated protein, were utilized to determine protein degradation. Furthermore, the SUnSET (surface sensing of translation) method was employed to determine rates of protein synthesis. RESULTS: MV treatment upregulated IGF-1 while downregulated FOXO1 and MURF1 (p < 0.05). AP administration reversed IGF-1, FOXO1 and MURF1 (p < 0.05), which was suppressed again by IGF-1 inhibition (p < 0.05), demonstrating the blockage of the IGF-1-FOXO1-MURF1 pathway. MV treatment caused decreased CMAP and cross-sectional areas of diaphragm muscle fibers, and increased time course of CMAP (p < 0.05). Additionally, oxidative stress, cell apoptosis, and protein degradation were increased and mitochondrial stability was decreased by MV treatment (p < 0.05). Conversely, AP administration reversed all these changes induced by MV, but this reversal was disrupted by the blockage of the IGF-1-FOXO1-MURF1 pathway. CONCLUSIONS: In this study, MV treatment induced symptoms of VIDD in rats, which were all effectively reversed by AP regulating the IGF-1-FOXO1-MURF1 pathway, demonstrating the potential of AP in ameliorating VIDD.


Subject(s)
Aminophylline , Diaphragm , Animals , Male , Rats , Aminophylline/pharmacology , Diaphragm/drug effects , Diaphragm/pathology , Diaphragm/physiopathology , Diaphragm/metabolism , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/genetics , Insulin-Like Growth Factor I/metabolism , Muscle Proteins/metabolism , Muscle Proteins/genetics , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Respiration, Artificial/adverse effects , Signal Transduction/drug effects , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics
14.
Nature ; 629(8010): 201-210, 2024 May.
Article in English | MEDLINE | ID: mdl-38600376

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapy has transformed the treatment of haematological malignancies such as acute lymphoblastic leukaemia, B cell lymphoma and multiple myeloma1-4, but the efficacy of CAR T cell therapy in solid tumours has been limited5. This is owing to a number of factors, including the immunosuppressive tumour microenvironment that gives rise to poorly persisting and metabolically dysfunctional T cells. Analysis of anti-CD19 CAR T cells used clinically has shown that positive treatment outcomes are associated with a more 'stem-like' phenotype and increased mitochondrial mass6-8. We therefore sought to identify transcription factors that could enhance CAR T cell fitness and efficacy against solid tumours. Here we show that overexpression of FOXO1 promotes a stem-like phenotype in CAR T cells derived from either healthy human donors or patients, which correlates with improved mitochondrial fitness, persistence and therapeutic efficacy in vivo. This work thus reveals an engineering approach to genetically enforce a favourable metabolic phenotype that has high translational potential to improve the efficacy of CAR T cells against solid tumours.


Subject(s)
Forkhead Box Protein O1 , Immunotherapy, Adoptive , Neoplasms , Receptors, Chimeric Antigen , Stem Cells , T-Lymphocytes , Humans , Mice , Cell Line, Tumor , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/genetics , Mitochondria/metabolism , Phenotype , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/cytology , Tumor Microenvironment/immunology , Stem Cells/cytology , Stem Cells/immunology , Stem Cells/metabolism , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy
15.
Nat Commun ; 15(1): 3440, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38653977

ABSTRACT

Oxidative stress from excess H2O2 activates transcription factors that restore redox balance and repair oxidative damage. Although many transcription factors are activated by H2O2, it is unclear whether they are activated at the same H2O2 concentration, or time. Dose-dependent activation is likely as oxidative stress is not a singular state and exhibits dose-dependent outcomes including cell-cycle arrest and cell death. Here, we show that transcription factor activation is both dose-dependent and coordinated over time. Low levels of H2O2 activate p53, NRF2 and JUN. Yet under high H2O2, these transcription factors are repressed, and FOXO1, NF-κB, and NFAT1 are activated. Time-lapse imaging revealed that the order in which these two groups of transcription factors are activated depends on whether H2O2 is administered acutely by bolus addition, or continuously through the glucose oxidase enzyme. Finally, we provide evidence that 2-Cys peroxiredoxins control which group of transcription factors are activated.


Subject(s)
Hydrogen Peroxide , Oxidative Stress , Transcription Factors , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Oxidative Stress/drug effects , Transcription Factors/metabolism , Transcription Factors/genetics , Humans , Peroxiredoxins/metabolism , Peroxiredoxins/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , NF-kappa B/metabolism , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/genetics , NFATC Transcription Factors/metabolism , Glucose Oxidase/metabolism , Animals
16.
Endocrinology ; 165(6)2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38679470

ABSTRACT

CONTEXT: Recurrent spontaneous abortion (RSA) is defined as the loss of 2 or more consecutive intrauterine pregnancies with the same sexual partner in the first trimester. Despite its significance, the etiology and underlying mechanisms of RSA remain elusive. Defective decidualization is proposed as one of the potential causes of RSA, with abnormal decidualization leading to disturbances in trophoblast invasion function. OBJECTIVE: To assess the role of bone morphogenetic protein 4 (BMP4) in decidualization and RSA. METHODS: Decidual samples were collected from both RSA patients and healthy controls to assess BMP4 expression. In vitro cell experiments utilized the hESC cell line to investigate the impact of BMP4 on decidualization and associated aging, as well as its role in the maternal-fetal interface communication. Subsequently, a spontaneous abortion mouse model was established to evaluate embryo resorption rates and BMP4 expression levels. RESULTS: Our study identified a significant downregulation of BMP4 expression in the decidua of RSA patients compared to the normal control group. In vitro, BMP4 knockdown resulted in inadequate decidualization and inhibited associated aging processes. Mechanistically, BMP4 was implicated in the regulation of FOXO1 expression, thereby influencing decidualization and aging. Furthermore, loss of BMP4 hindered trophoblast migration and invasion via FOXO1 modulation. Additionally, BMP4 downregulation was observed in RSA mice. CONCLUSION: Our findings highlighted the downregulation of BMP4 in both RSA patients and mice. BMP4 in human endometrial stromal cells was shown to modulate decidualization by regulating FOXO1 expression. Loss of BMP4 may contribute to the pathogenesis of RSA, suggesting potential avenues for abortion prevention strategies.


Subject(s)
Abortion, Habitual , Bone Morphogenetic Protein 4 , Decidua , Endometrium , Forkhead Box Protein O1 , Stromal Cells , Female , Humans , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 4/genetics , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/genetics , Stromal Cells/metabolism , Animals , Mice , Decidua/metabolism , Pregnancy , Endometrium/metabolism , Endometrium/cytology , Abortion, Habitual/metabolism , Abortion, Habitual/genetics , Adult , Trophoblasts/metabolism , Case-Control Studies
17.
Nat Commun ; 15(1): 2859, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38570500

ABSTRACT

Cold-induced injuries severely limit opportunities and outcomes of hypothermic therapies and organ preservation, calling for better understanding of cold adaptation. Here, by surveying cold-altered chromatin accessibility and integrated CUT&Tag/RNA-seq analyses in human stem cells, we reveal forkhead box O1 (FOXO1) as a key transcription factor for autonomous cold adaptation. Accordingly, we find a nonconventional, temperature-sensitive FOXO1 transport mechanism involving the nuclear pore complex protein RANBP2, SUMO-modification of transporter proteins Importin-7 and Exportin-1, and a SUMO-interacting motif on FOXO1. Our conclusions are supported by cold survival experiments with human cell models and zebrafish larvae. Promoting FOXO1 nuclear entry by the Exportin-1 inhibitor KPT-330 enhances cold tolerance in pre-diabetic obese mice, and greatly prolongs the shelf-life of human and mouse pancreatic tissues and islets. Transplantation of mouse islets cold-stored for 14 days reestablishes normoglycemia in diabetic mice. Our findings uncover a regulatory network and potential therapeutic targets to boost spontaneous cold adaptation.


Subject(s)
Diabetes Mellitus, Experimental , Forkhead Transcription Factors , Mice , Humans , Animals , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Active Transport, Cell Nucleus , Zebrafish/metabolism , Karyopherins/metabolism
18.
Am J Dermatopathol ; 46(4): 223-227, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38457669

ABSTRACT

ABSTRACT: Epithelioid hemangioma (EH) is a benign vascular tumor displaying diverse histomorphologies. Among these, one EH subtype comprises cellular sheets of atypical epithelioid cells, posing potential challenges in distinguishing it from malignant vascular lesions. In this case report, we present a cutaneous cellular EH that carries the rare GATA6::FOXO1 gene fusion, a recent discovery. Our aim is to provide an updated insight into the evolving knowledge of EHs while delving into the histologic and molecular characteristics of the primary differential diagnoses.


Subject(s)
Angiolymphoid Hyperplasia with Eosinophilia , Hemangioendothelioma, Epithelioid , Hemangioma , Vascular Neoplasms , Humans , Angiolymphoid Hyperplasia with Eosinophilia/pathology , Hemangioma/pathology , Gene Fusion , Diagnosis, Differential , Hemangioendothelioma, Epithelioid/genetics , Forkhead Box Protein O1/genetics , GATA6 Transcription Factor/genetics
19.
J Biol Chem ; 300(4): 107209, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38519029

ABSTRACT

FOXO1 is a transcription factor and potential tumor suppressor that is negatively regulated downstream of PI3K-PKB/AKT signaling. Paradoxically, FOXO also promotes tumor growth, but the detailed mechanisms behind this role of FOXO are not fully understood. In this study, we revealed a molecular cascade by which the Thr24 residue of FOXO1 is phosphorylated by AKT and is dephosphorylated by calcineurin, which is a Ca2+-dependent protein phosphatase. Curiously, single nucleotide somatic mutations of FOXO1 in cancer occur frequently at and near Thr24. Using a calcineurin inhibitor and shRNA directed against calcineurin, we revealed that calcineurin-mediated dephosphorylation of Thr24 regulates FOXO1 protein stability. We also found that FOXO1 binds to the promoter region of MDM2 and activates transcription, which in turn promotes MDM2-mediated ubiquitination and degradation of p53. FOXO3a and FOXO4 are shown to control p53 activity; however, the significance of FOXO1 in p53 regulation remains largely unknown. Supporting this notion, FOXO1 depletion increased p53 and p21 protein levels in association with the inhibition of cell proliferation. Taken together, these results indicate that FOXO1 is stabilized by calcineurin-mediated dephosphorylation and that FOXO1 supports cancer cell proliferation by promoting MDM2 transcription and subsequent p53 degradation.


Subject(s)
Calcineurin , Cell Proliferation , Forkhead Box Protein O1 , Proteolysis , Proto-Oncogene Proteins c-mdm2 , Tumor Suppressor Protein p53 , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins c-mdm2/genetics , Humans , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/genetics , Calcineurin/metabolism , Calcineurin/genetics , Phosphorylation , Ubiquitination , Cell Line, Tumor , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/genetics , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/genetics , Protein Stability
20.
J Physiol Biochem ; 80(2): 381-392, 2024 May.
Article in English | MEDLINE | ID: mdl-38536659

ABSTRACT

Hepatocellular carcinoma (HCC) is a highly vascularized tumor, one of the most common and lethal cancer-related tumor deaths worldwide, with cell proliferation playing a key role. In this study our western blot results and data from TAGC demonstrate a strong association between Sorcin (SRI) overexpression and poor outcomes in HCC. Moreover, SRI overexpression was remarkably effective in promoting proliferation in vitro and increasing tumor growth in vivo, which were attenuated by knocking down SRI. Mechanistically, SRI regulated vascular endothelial growth factor A (VEGFA) and vascular endothelial growth factor B (VEGFB) through PI3K/Akt/FOXO1 signal pathway. Overall, our study indicates that SRI stimulates HCC growth by controlling VEGFA/B, which presents a fresh insight into the pathogenesis of hepatocarcinogenesis and a new therapeutic target for HCC.


Subject(s)
Calcium-Binding Proteins , Carcinoma, Hepatocellular , Cell Proliferation , Liver Neoplasms , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor B , Animals , Humans , Mice , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/genetics , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/genetics , Mice, Inbred BALB C , Mice, Nude , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor B/metabolism , Vascular Endothelial Growth Factor B/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...