Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 853
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 19(9): 986-1000, 2018 09.
Article in English | MEDLINE | ID: mdl-30127432

ABSTRACT

Gain-of-function mutations in the gene encoding the phosphatidylinositol-3-OH kinase catalytic subunit p110δ (PI3Kδ) result in a human primary immunodeficiency characterized by lymphoproliferation, respiratory infections and inefficient responses to vaccines. However, what promotes these immunological disturbances at the cellular and molecular level remains unknown. We generated a mouse model that recapitulated major features of this disease and used this model and patient samples to probe how hyperactive PI3Kδ fosters aberrant humoral immunity. We found that mutant PI3Kδ led to co-stimulatory receptor ICOS-independent increases in the abundance of follicular helper T cells (TFH cells) and germinal-center (GC) B cells, disorganized GCs and poor class-switched antigen-specific responses to immunization, associated with altered regulation of the transcription factor FOXO1 and pro-apoptotic and anti-apoptotic members of the BCL-2 family. Notably, aberrant responses were accompanied by increased reactivity to gut bacteria and a broad increase in autoantibodies that were dependent on stimulation by commensal microbes. Our findings suggest that proper regulation of PI3Kδ is critical for ensuring optimal host-protective humoral immunity despite tonic stimulation from the commensal microbiome.


Subject(s)
B-Lymphocytes/physiology , Gastrointestinal Microbiome/immunology , Germinal Center/physiology , Mutation/genetics , Phosphatidylinositol 3-Kinases/genetics , T-Lymphocytes, Helper-Inducer/physiology , Animals , Autoantibodies/blood , Cells, Cultured , Class I Phosphatidylinositol 3-Kinases/genetics , Disease Models, Animal , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Humans , Immunity, Humoral/genetics , Immunoglobulin Class Switching/genetics , Immunologic Deficiency Syndromes/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism
2.
Nat Immunol ; 19(8): 838-848, 2018 08.
Article in English | MEDLINE | ID: mdl-29988091

ABSTRACT

Foxo transcription factors play an essential role in regulating specialized lymphocyte functions and in maintaining T cell quiescence. Here, we used a system in which Foxo1 transcription-factor activity, which is normally terminated upon cell activation, cannot be silenced, and we show that enforcing Foxo1 activity disrupts homeostasis of CD4 conventional and regulatory T cells. Despite limiting cell metabolism, continued Foxo1 activity is associated with increased activation of the kinase Akt and a cell-intrinsic proliferative advantage; however, survival and cell division are decreased in a competitive setting or growth-factor-limiting conditions. Via control of expression of the transcription factor Myc and the IL-2 receptor ß-chain, termination of Foxo1 signaling couples the increase in cellular cholesterol to biomass accumulation after activation, thereby facilitating immunological synapse formation and mTORC1 activity. These data reveal that Foxo1 regulates the integration of metabolic and mitogenic signals essential for T cell competitive fitness and the coordination of cell growth with cell division.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Forkhead Box Protein O1/metabolism , T-Lymphocytes, Regulatory/physiology , Animals , Cell Proliferation , Cells, Cultured , Cholesterol/metabolism , Forkhead Box Protein O1/genetics , Gene Expression Profiling , Homeostasis , Immunological Synapses/metabolism , Interleukin-2 Receptor beta Subunit/genetics , Interleukin-2 Receptor beta Subunit/metabolism , Lymphocyte Activation , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Knockout , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction
3.
Immunity ; 54(8): 1807-1824.e14, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34380064

ABSTRACT

The transcription factor forkhead box O1 (FOXO1), which instructs the dark zone program to direct germinal center (GC) polarity, is typically inactivated by phosphatidylinositol 3-kinase (PI3K) signals. Here, we investigated how FOXO1 mutations targeting this regulatory axis in GC-derived B cell non-Hodgkin lymphomas (B-NHLs) contribute to lymphomagenesis. Examination of primary B-NHL tissues revealed that FOXO1 mutations and PI3K pathway activity were not directly correlated. Human B cell lines bearing FOXO1 mutations exhibited hyperactivation of PI3K and Stress-activated protein kinase (SAPK)/Jun amino-terminal kinase (JNK) signaling, and increased cell survival under stress conditions as a result of alterations in FOXO1 transcriptional affinities and activation of transcriptional programs characteristic of GC-positive selection. When modeled in mice, FOXO1 mutations conferred competitive advantage to B cells in response to key T-dependent immune signals, disrupting GC homeostasis. FOXO1 mutant transcriptional signatures were prevalent in human B-NHL and predicted poor clinical outcomes. Thus, rather than enforcing FOXO1 constitutive activity, FOXO1 mutations enable co-option of GC-positive selection programs during the pathogenesis of GC-derived lymphomas.


Subject(s)
B-Lymphocytes/cytology , Forkhead Box Protein O1/genetics , Germinal Center/immunology , Lymphoma, B-Cell/pathology , Animals , B-Lymphocytes/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Line , Cell Proliferation/genetics , Cell Survival/genetics , Gene Expression Regulation/genetics , HEK293 Cells , Humans , Lymphoma, B-Cell/genetics , MAP Kinase Kinase 4/metabolism , Mice , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/genetics , Signal Transduction/immunology
4.
Nature ; 630(8018): 976-983, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38867048

ABSTRACT

Interleukin (IL-)23 is a major mediator and therapeutic target in chronic inflammatory diseases that also elicits tissue protection in the intestine at homeostasis or following acute infection1-4. However, the mechanisms that shape these beneficial versus pathological outcomes remain poorly understood. To address this gap in knowledge, we performed single-cell RNA sequencing on all IL-23 receptor-expressing cells in the intestine and their acute response to IL-23, revealing a dominance of T cells and group 3 innate lymphoid cells (ILC3s). Unexpectedly, we identified potent upregulation of the immunoregulatory checkpoint molecule cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) on ILC3s. This pathway was activated by gut microbes and IL-23 in a FOXO1- and STAT3-dependent manner. Mice lacking CTLA-4 on ILC3s exhibited reduced regulatory T cells, elevated inflammatory T cells and more-severe intestinal inflammation. IL-23 induction of CTLA-4+ ILC3s was necessary and sufficient to reduce co-stimulatory molecules and increase PD-L1 bioavailability on intestinal myeloid cells. Finally, human ILC3s upregulated CTLA-4 in response to IL-23 or gut inflammation and correlated with immunoregulation in inflammatory bowel disease. These results reveal ILC3-intrinsic CTLA-4 as an essential checkpoint that restrains the pathological outcomes of IL-23, suggesting that disruption of these lymphocytes, which occurs in inflammatory bowel disease5-7, contributes to chronic inflammation.


Subject(s)
CTLA-4 Antigen , Immunity, Innate , Inflammation , Interleukin-23 , Lymphocytes , Animals , CTLA-4 Antigen/metabolism , Mice , Interleukin-23/metabolism , Inflammation/immunology , Inflammation/pathology , Inflammation/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Female , Male , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , STAT3 Transcription Factor/metabolism , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/genetics , B7-H1 Antigen/metabolism , Gastrointestinal Microbiome , Mice, Inbred C57BL , Intestines/immunology , Intestines/pathology , Single-Cell Analysis , Humans , RNA-Seq
5.
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
6.
Nature ; 598(7881): 473-478, 2021 10.
Article in English | MEDLINE | ID: mdl-34646017

ABSTRACT

The progression of chronic liver disease to hepatocellular carcinoma is caused by the acquisition of somatic mutations that affect 20-30 cancer genes1-8. Burdens of somatic mutations are higher and clonal expansions larger in chronic liver disease9-13 than in normal liver13-16, which enables positive selection to shape the genomic landscape9-13. Here we analysed somatic mutations from 1,590 genomes across 34 liver samples, including healthy controls, alcohol-related liver disease and non-alcoholic fatty liver disease. Seven of the 29 patients with liver disease had mutations in FOXO1, the major transcription factor in insulin signalling. These mutations affected a single hotspot within the gene, impairing the insulin-mediated nuclear export of FOXO1. Notably, six of the seven patients with FOXO1S22W hotspot mutations showed convergent evolution, with variants acquired independently by up to nine distinct hepatocyte clones per patient. CIDEB, which regulates lipid droplet metabolism in hepatocytes17-19, and GPAM, which produces storage triacylglycerol from free fatty acids20,21, also had a significant excess of mutations. We again observed frequent convergent evolution: up to fourteen independent clones per patient with CIDEB mutations and up to seven clones per patient with GPAM mutations. Mutations in metabolism genes were distributed across multiple anatomical segments of the liver, increased clone size and were seen in both alcohol-related liver disease and non-alcoholic fatty liver disease, but rarely in hepatocellular carcinoma. Master regulators of metabolic pathways are a frequent target of convergent somatic mutation in alcohol-related and non-alcoholic fatty liver disease.


Subject(s)
Liver Diseases/genetics , Liver Diseases/metabolism , Liver/metabolism , Mutation/genetics , Active Transport, Cell Nucleus/genetics , Apoptosis Regulatory Proteins/genetics , Cell Line, Tumor , Chronic Disease , Cohort Studies , Fatty Acids, Nonesterified/metabolism , Female , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Humans , Insulin Resistance , Liver Diseases, Alcoholic/genetics , Liver Diseases, Alcoholic/metabolism , Male , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Triglycerides/metabolism
7.
Mol Cell ; 75(4): 807-822.e8, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31442424

ABSTRACT

mTORC2 controls glucose and lipid metabolism, but the mechanisms are unclear. Here, we show that conditionally deleting the essential mTORC2 subunit Rictor in murine brown adipocytes inhibits de novo lipid synthesis, promotes lipid catabolism and thermogenesis, and protects against diet-induced obesity and hepatic steatosis. AKT kinases are the canonical mTORC2 substrates; however, deleting Rictor in brown adipocytes appears to drive lipid catabolism by promoting FoxO1 deacetylation independently of AKT, and in a pathway distinct from its positive role in anabolic lipid synthesis. This facilitates FoxO1 nuclear retention, enhances lipid uptake and lipolysis, and potentiates UCP1 expression. We provide evidence that SIRT6 is the FoxO1 deacetylase suppressed by mTORC2 and show an endogenous interaction between SIRT6 and mTORC2 in both mouse and human cells. Our findings suggest a new paradigm of mTORC2 function filling an important gap in our understanding of this more mysterious mTOR complex.


Subject(s)
Adipocytes, Brown/metabolism , Forkhead Box Protein O1/metabolism , Lipolysis , Mechanistic Target of Rapamycin Complex 2/metabolism , Sirtuins/metabolism , Adipocytes, Brown/cytology , Animals , Forkhead Box Protein O1/genetics , HEK293 Cells , HeLa Cells , Humans , Mechanistic Target of Rapamycin Complex 2/genetics , Mice , Mice, Transgenic , Rapamycin-Insensitive Companion of mTOR Protein/genetics , Rapamycin-Insensitive Companion of mTOR Protein/metabolism , Sirtuins/genetics
8.
Immunity ; 46(4): 596-608, 2017 04 18.
Article in English | MEDLINE | ID: mdl-28410989

ABSTRACT

Understanding immunological memory formation depends on elucidating how multipotent memory precursor (MP) cells maintain developmental plasticity and longevity to provide long-term immunity while other effector cells develop into terminally differentiated effector (TE) cells with limited survival. Profiling active (H3K27ac) and repressed (H3K27me3) chromatin in naive, MP, and TE CD8+ T cells during viral infection revealed increased H3K27me3 deposition at numerous pro-memory and pro-survival genes in TE relative to MP cells, indicative of fate restriction, but permissive chromatin at both pro-memory and pro-effector genes in MP cells, indicative of multipotency. Polycomb repressive complex 2 deficiency impaired clonal expansion and TE cell differentiation, but minimally impacted CD8+ memory T cell maturation. Abundant H3K27me3 deposition at pro-memory genes occurred late during TE cell development, probably from diminished transcription factor FOXO1 expression. These results outline a temporal model for loss of memory cell potential through selective epigenetic silencing of pro-memory genes in effector T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Chromatin/immunology , Polycomb Repressive Complex 2/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Chromatin/genetics , Chromatin/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/immunology , Enhancer of Zeste Homolog 2 Protein/metabolism , Flow Cytometry , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/immunology , Forkhead Box Protein O1/metabolism , Gene Expression/immunology , Histones/immunology , Histones/metabolism , Immunoblotting , Immunologic Memory/genetics , Immunologic Memory/immunology , Lysine/immunology , Lysine/metabolism , Methylation , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Immunological , Multipotent Stem Cells/immunology , Multipotent Stem Cells/metabolism , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Reverse Transcriptase Polymerase Chain Reaction
9.
Immunity ; 47(6): 1067-1082.e12, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29246441

ABSTRACT

Roquin proteins preclude spontaneous T cell activation and aberrant differentiation of T follicular helper (Tfh) or T helper 17 (Th17) cells. Here we showed that deletion of Roquin-encoding alleles specifically in regulatory T (Treg) cells also caused the activation of conventional T cells. Roquin-deficient Treg cells downregulated CD25, acquired a follicular Treg (Tfr) cell phenotype, and suppressed germinal center reactions but could not protect from colitis. Roquin inhibited the PI3K-mTOR signaling pathway by upregulation of Pten through interfering with miR-17∼92 binding to an overlapping cis-element in the Pten 3' UTR, and downregulated the Foxo1-specific E3 ubiquitin ligase Itch. Loss of Roquin enhanced Akt-mTOR signaling and protein synthesis, whereas inhibition of PI3K or mTOR in Roquin-deficient T cells corrected enhanced Tfh and Th17 or reduced iTreg cell differentiation. Thereby, Roquin-mediated control of PI3K-mTOR signaling prevents autoimmunity by restraining activation and differentiation of conventional T cells and specialization of Treg cells.


Subject(s)
Colitis/immunology , Phosphatidylinositol 3-Kinases/immunology , Repressor Proteins/immunology , TOR Serine-Threonine Kinases/immunology , Ubiquitin-Protein Ligases/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Differentiation , Colitis/genetics , Colitis/pathology , Disease Models, Animal , Female , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/immunology , Gene Expression Regulation , Germinal Center/immunology , Germinal Center/pathology , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/genetics , MicroRNAs/immunology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/immunology , Phosphatidylinositol 3-Kinases/genetics , Primary Cell Culture , Repressor Proteins/deficiency , Repressor Proteins/genetics , Signal Transduction , Spleen/immunology , Spleen/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , TOR Serine-Threonine Kinases/genetics , Th17 Cells/immunology , Th17 Cells/pathology , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics
10.
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
11.
Development ; 149(7)2022 04 01.
Article in English | MEDLINE | ID: mdl-35297995

ABSTRACT

Establishing a functional circulatory system is required for post-implantation development during murine embryogenesis. Previous studies in loss-of-function mouse models showed that FOXO1, a Forkhead family transcription factor, is required for yolk sac (YS) vascular remodeling and survival beyond embryonic day (E) 11. Here, we demonstrate that at E8.25, loss of Foxo1 in Tie2-cre expressing cells resulted in increased sprouty 2 (Spry2) and Spry4 expression, reduced arterial gene expression and reduced Kdr (also known as Vegfr2 and Flk1) transcripts without affecting overall endothelial cell identity, survival or proliferation. Using a Dll4-BAC-nlacZ reporter line, we found that one of the earliest expressed arterial genes, delta like 4, is significantly reduced in Foxo1 mutant YS without being substantially affected in the embryo proper. We show that FOXO1 binds directly to previously identified Spry2 gene regulatory elements (GREs) and newly identified, evolutionarily conserved Spry4 GREs to repress their expression. Furthermore, overexpression of Spry4 in transient transgenic embryos largely recapitulates the reduced expression of arterial genes seen in conditional Foxo1 mutants. Together, these data reveal a novel role for FOXO1 as a key transcriptional repressor regulating both pre-flow arterial specification and subsequent vessel remodeling within the murine YS.


Subject(s)
Nerve Tissue Proteins/metabolism , Vascular Remodeling , Yolk Sac , Animals , Arteries , Embryo, Mammalian/metabolism , Endothelial Cells/metabolism , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Mice , Vascular Remodeling/genetics , Yolk Sac/metabolism
12.
Immunity ; 44(6): 1284-98, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27332731

ABSTRACT

T helper 17 (Th17) cells are key players in autoimmune diseases. However, the roles of non-coding RNAs in Th17 cell development and function are largely unknown. We found that deletion of the endoribonuclease-encoding Dicer1 specifically in Th17 cells protected mice from experimental autoimmune encephalomyelitis. We found that the Dicer1-regulated microRNA (miR)-183-96-182 cluster (miR-183C) was highly expressed in Th17 cells and was induced by cytokine IL-6-STAT3 signaling. miR-183C expression enhanced pathogenic cytokine production from Th17 cells during their development and promoted autoimmunity. Mechanistically, miR-183C in Th17 cells directly repressed expression of the transcription factor Foxo1. Foxo1 negatively regulated the pathogenicity of Th17 cells in part by inhibiting expression of cytokine receptor IL-1R1. These findings indicate that the miR-183C drives Th17 pathogenicity in autoimmune diseases via inhibition of Foxo1 and present promising therapeutic targets.


Subject(s)
DEAD-box RNA Helicases/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Forkhead Box Protein O1/metabolism , MicroRNAs/genetics , Multiple Sclerosis/immunology , Ribonuclease III/metabolism , Th17 Cells/physiology , Animals , Cells, Cultured , DEAD-box RNA Helicases/genetics , Forkhead Box Protein O1/genetics , Humans , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-1 Type I/metabolism , Ribonuclease III/genetics , STAT3 Transcription Factor/metabolism
13.
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
14.
PLoS Genet ; 18(5): e1009782, 2022 05.
Article in English | MEDLINE | ID: mdl-35604932

ABSTRACT

The hallmarks of the alveolar subclass of rhabdomyosarcoma are chromosomal translocations that generate chimeric PAX3-FOXO1 or PAX7-FOXO1 transcription factors. Overexpression of either PAX-FOXO1s results in related cell transformation in animal models. Yet, in patients the two structural genetic aberrations they derived from are associated with distinct pathological manifestations. To assess the mechanisms underlying these differences, we generated isogenic fibroblast lines expressing either PAX-FOXO1 paralog. Mapping of their genomic recruitment using CUT&Tag revealed that the two chimeric proteins have distinct DNA binding preferences. In addition, PAX7-FOXO1 binding results in greater recruitment of the H3K27ac activation mark than PAX3-FOXO1 binding and is accompanied by greater transcriptional activation of neighbouring genes. These effects are associated with a PAX-FOXO1-specific alteration in the expression of genes regulating cell shape and the cell cycle. Consistently, PAX3-FOXO1 accentuates fibroblast cellular traits associated with contractility and surface adhesion and limits entry into S phase. In contrast, PAX7-FOXO1 drives cells to adopt an amoeboid shape, reduces entry into M phase, and causes increased DNA damage. Altogether, our results argue that the diversity of rhabdomyosarcoma manifestation arises, in part, from the divergence between the genomic occupancy and transcriptional activity of PAX3-FOXO1 and PAX7-FOXO1.


Subject(s)
Oncogene Proteins, Fusion , Paired Box Transcription Factors , Rhabdomyosarcoma, Alveolar , Animals , Cell Line , Cell Transformation, Neoplastic/genetics , Fibroblasts , Forkhead Box Protein O1/genetics , Forkhead Transcription Factors/genetics , Humans , Oncogene Proteins, Fusion/genetics , PAX3 Transcription Factor/genetics , PAX7 Transcription Factor/genetics , Paired Box Transcription Factors/genetics , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma, Alveolar/genetics
15.
Lancet Oncol ; 25(7): 912-921, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38936378

ABSTRACT

BACKGROUND: The Children's Oncology Group defines intermediate-risk rhabdomyosarcoma as unresected FOXO1 fusion-negative disease arising at an unfavourable site or non-metastatic FOXO1 fusion-positive disease. Temsirolimus in combination with chemotherapy has shown promising activity in patients with relapsed or refractory rhabdomyosarcoma. We aimed to compare event-free survival in patients with intermediate-risk rhabdomyosarcoma treated with vincristine, actinomycin, and cyclophosphamide alternating with vincristine and irinotecan (VAC/VI) combined with temsirolimus followed by maintenance therapy versus VAC/VI alone with maintenance therapy. METHODS: ARST1431 was a randomised, open-label, phase 3 trial conducted across 210 institutions in Australia, Canada, New Zealand, and the USA. Eligible patients were those aged 40 years or younger with non-metastatic FOXO1-positive rhabdomyosarcoma or unresected FOXO1-negative rhabdomyosarcoma disease from unfavourable sites. Two other groups of patients were also eligible: those who had FOXO1-negative disease at a favourable site (excluding orbit) that was unresected; and those who were aged younger than 10 years with stage IV FOXO1-negative disease with distant metastases. Eligible patients had to have a Lansky performance status score of 50 or higher if 16 years or younger and a Karnofsky performance status score of 50 or higher if older than 16 years; all patients were previously untreated. Patients were randomised (1:1) in blocks of four and stratified by histology, stage, and group. Patients received intravenous VAC/VI chemotherapy with a cyclophosphamide dose of 1·2 g/m2 per dose per cycle with or without a reducing dose of intravenous weekly temsirolimus starting at 15 mg/m2 or 0·5 mg/kg per dose for those who weighed less than 10 kg. The total duration of therapy was 42 weeks followed by 6 months of maintenance therapy with oral cyclophosphamide plus intravenous vinorelbine for all patients. Temsirolimus was withheld during radiotherapy and for 2 weeks before any major surgical procedure. The primary endpoint was 3-year event-free survival. Data were analysed with a revised intention-to-treat approach. The study is registered with ClinicalTrials.gov (NCT02567435) and is complete. FINDINGS: Between May 23, 2016, and Jan 1, 2022, 325 patients were enrolled. In 297 evaluable patients (148 assigned to VAC/VI alone and 149 assigned to VAC/VI with temsirolimus), the median age was 6·3 years (IQR 3·0-11·3); 33 (11%) patients were aged 18 years or older; 179 (60%) of 297 were male. 113 (77%) of 148 patients were FOXO1 negative in the VAC/VI group, and 108 (73%) of 149 were FOXO1 negative in the VAC/VI with temsirolimus group. With a median follow-up of 3·6 years (IQR 2·8-4·5), 3-year event-free survival did not differ significantly between the two groups (64·8% [95% CI 55·5-74·1] in the VAC/VI group vs 66·8% [57·5-76·2] in the VAC/VI plus temsirolimus group (hazard ratio 0·86 [95% CI 0·58-1·26]; log-rank p=0·44). The most common grade 3-4 adverse events were anaemia (62 events in 60 [41%] of 148 patients in the VAC/VI group vs 89 events in 87 [58%] of 149 patients in the VAC/VI with temsirolimus group), lymphopenia (83 events in 65 [44%] vs 99 events in 71 [48%]), neutropenia (160 events in 99 [67%] vs 164 events in 105 [70%]), and leukopenia (121 events in 86 [58%] vs 132 events in 93 [62%]). There was one treatment-related death in the VAC/VI with temsirolimus group, categorised as not otherwise specified. INTERPRETATION: Addition of temsirolimus to VAC/VI did not improve event-free survival in patients with intermediate-risk rhabdomyosarcoma defined by their FOXO1 translocation status and clinical factors. Novel biology-based strategies are needed to improve outcomes in this population. FUNDING: The Children's Oncology Group (supported by the US National Cancer Institute, US National Institutes of Health).


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Cyclophosphamide , Rhabdomyosarcoma , Sirolimus , Vincristine , Humans , Male , Female , Child , Adolescent , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Sirolimus/analogs & derivatives , Sirolimus/administration & dosage , Sirolimus/therapeutic use , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/mortality , Rhabdomyosarcoma/pathology , Child, Preschool , Vincristine/administration & dosage , Vincristine/adverse effects , Young Adult , Cyclophosphamide/administration & dosage , Adult , Dactinomycin/administration & dosage , Irinotecan/administration & dosage , Irinotecan/therapeutic use , Infant , Progression-Free Survival , Forkhead Box Protein O1/genetics
16.
Am J Physiol Lung Cell Mol Physiol ; 326(3): L344-L352, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38252663

ABSTRACT

We have discovered intrinsically fibrogenic mesenchymal progenitor cells (MPCs) in the human idiopathic pulmonary fibrosis (IPF) lung. IPF MPCs display a durably distinct transcriptome, suggesting that they have undergone epigenetic modifications. Prior studies indicate that the chromatin remodeler Brg1 associates with the arginine methyltransferase PRMT5 to epigenetically regulate transcription factors. We hypothesize that a Brg1/PRMT5 nuclear complex epigenetically regulates critical nodes in IPF MPC self-renewal signaling networks. IPF and control MPCs were isolated from primary mesenchymal cell lines established from IPF and control patients. RNA-sequencing identified increased expression of the FOXO1 transcription factor in IPF MPCs compared with controls, a result we confirmed by Q-PCR and Western blot analysis. Immunoprecipitation identified a CD44/Brg1/PRMT5 nuclear complex in IPF MPCs. Chromatin immunoprecipitation assays showed that PRMT5 and its methylation mark H3R2me2 are enriched on the FOXO1 promoter. We show that loss of Brg1 and PRMT5 function decreases FOXO1 expression and impairs IPF MPC self-renewal, and that loss of FOXO1 function decreases IPF MPC self-renewal and expression of the SOX2 and OCT4 stemness markers. Our findings indicate that the FOXO1 gene is overexpressed in IPF MPCs in a CD44/Brg1/PRMT5 nuclear complex-dependent manner. Our data suggest that Brg1 alters chromatin accessibility, enriching PRMT5 occupancy on the FOXO1 promoter, and PRMT5 methylates histone H3 arginine 2 (H3R2) on the FOXO1 promoter, increasing its expression. Our data are in accord with the concept that this coordinated interplay is responsible for promoting IPF MPC self-renewal and maintaining a critical pool of fibrogenic MPCs that drive IPF progression.NEW & NOTEWORTHY Our research offers valuable understanding regarding the epigenetic control of IPF MPC. The data we obtained strongly support the idea that the coordination between chromatin remodeling and histone methylation plays a key role in regulating transcription factors. Specifically, our findings indicate that FOXO1, an essential transcription factor, likely governs the self-renewal of IPF MPC, which is crucial for maintaining a critical pool of fibrogenic MPCs. This interplay could be an important therapeutic target.


Subject(s)
Idiopathic Pulmonary Fibrosis , Mesenchymal Stem Cells , Humans , Transcription Factors/genetics , Transcription Factors/metabolism , Gene Expression Regulation , Histones/metabolism , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/metabolism , Chromatin/metabolism , Mesenchymal Stem Cells/metabolism , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism
17.
Cancer Sci ; 115(4): 1224-1240, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38403332

ABSTRACT

The transcription factor forkhead box protein O1 (FoxO1) is closely related to the occurrence and development of ovarian cancer (OC), however its role and molecular mechanisms remain unclear. Herein, we found that FoxO1 was highly expressed in clinical samples of OC patients and was significantly correlated with poor prognosis. FoxO1 knockdown inhibited the proliferation of OC cells in vitro and in vivo. ChIP-seq combined with GEPIA2 and Kaplan-Meier database analysis showed that structural maintenance of chromosome 4 (SMC4) is a downstream target of FoxO1, and FoxO1 promotes SMC4 transcription by binding to its -1400/-1390 bp promoter. The high expression of SMC4 significantly blocked the tumor inhibition effect of FoxO1 knockdown. Furtherly, FoxO1 increased SMC4 mRNA abundance by transcriptionally activating methyltransferase-like 14 (METTL14) and increasing SMC4 m6A methylation on its coding sequence region. The Cancer Genome Atlas dataset analysis confirmed a significant positive correlation between FoxO1, SMC4, and METTL14 expression in OC. In summary, this study revealed the molecular mechanisms of FoxO1 regulating SMC4 and established a clinical link between the expression of FoxO1/METTL14/SMC4 in the occurrence of OC, thus providing a potential diagnostic target and therapeutic strategy.


Subject(s)
Chromosomes, Human, Pair 4 , Ovarian Neoplasms , Female , Humans , Adenosine Triphosphatases/genetics , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/genetics , Chromosomes, Human, Pair 4/metabolism , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Kaplan-Meier Estimate , Methyltransferases/genetics , Ovarian Neoplasms/pathology
18.
Cancer ; 130(13): 2351-2360, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38400828

ABSTRACT

BACKGROUND: The objective of this study was to investigate the role of clinical factors together with FOXO1 fusion status in patients with nonmetastatic rhabdomyosarcoma (RMS) to develop a predictive model for event-free survival and provide a rationale for risk stratification in future trials. METHODS: The authors used data from patients enrolled in the European Pediatric Soft Tissue Sarcoma Study Group (EpSSG) RMS 2005 study (EpSSG RMS 2005; EudraCT number 2005-000217-35). The following baseline variables were considered for the multivariable model: age at diagnosis, sex, histology, primary tumor site, Intergroup Rhabdomyosarcoma Studies group, tumor size, nodal status, and FOXO1 fusion status. Main effects and significant second-order interactions of candidate predictors were included in a multiple Cox proportional hazards regression model. A nomogram was generated for predicting 5-year event-free survival (EFS) probabilities. RESULTS: The EFS and overall survival rates at 5 years were 70.9% (95% confidence interval, 68.6%-73.1%) and 81.0% (95% confidence interval, 78.9%-82.8%), respectively. The multivariable model retained five prognostic factors, including age at diagnosis interacting with tumor size, tumor primary site, Intergroup Rhabdomyosarcoma Studies clinical group, and FOXO1 fusion status. Based on each patient's total score in the nomogram, patients were stratified into four groups. The 5-year EFS rates were 94.1%, 78.4%, 65.2%, and 52.1% in the low-risk, intermediate-risk, high-risk, and very-high-risk groups, respectively, and the corresponding 5-year overall survival rates were 97.2%, 91.5%, 74.3%, and 60.8%, respectively. CONCLUSIONS: The results presented here provide the rationale to modify the EpSSG stratification, with the most significant change represented by the replacement of histology with fusion status. This classification was adopted in the new international trial launched by the EpSSG.


Subject(s)
Nomograms , Rhabdomyosarcoma , Humans , Rhabdomyosarcoma/mortality , Rhabdomyosarcoma/pathology , Rhabdomyosarcoma/therapy , Male , Female , Child, Preschool , Child , Prognosis , Infant , Risk Assessment , Adolescent , Europe/epidemiology , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Oncogene Proteins, Fusion/genetics
19.
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
20.
Funct Integr Genomics ; 24(1): 6, 2024 Jan 08.
Article in English | MEDLINE | ID: mdl-38189995

ABSTRACT

The aim of this study was to explore the role of forkhead box transcription Factor O1 (FoxO1) in chronic inflammation in polycystic ovary syndrome (PCOS). A PCOS rat model was constructed as an in vivo model by letrozole induction, and granulosa cells (GCs) from PCOS rats were isolated and cultured as an in vitro cellular model. FoxO1 was knocked down by shRNA and siRNA in the PCOS rat model and GCs model, respectively. H&E staining was conducted to evaluate the effect of FoxO1 inhibition on ovarian pathology and dysfunction in PCOS rats. The levels of inflammatory cytokines in the ovaries and uterus of PCOS rats and in GCs were assessed by ELISA. Flow cytometry was used to evaluate the changes in the contents of neutrophils and macrophages in the peripheral blood and spleen of PCOS rats. CCK-8 assays and Annexin V-FITC/PI staining were performed to evaluate the proliferation and apoptosis of GCs. The expression of genes and proteins related to the TLR4/NF-κB/NLRP3 pathway in GCs was determined by RT-qPCR and Western blotting. The results indicated that FoxO1 was highly expressed in PCOS rat model. Inhibition of FoxO1 significantly mitigated the pathological changes and dysfunction in the ovaries of PCOS rats while also suppressing inflammation and fibrosis in the ovaries and uterus. Moreover, knocking down FoxO1 facilitated the restoration of the normal ratio of neutrophils and macrophages in the peripheral blood and spleen of PCOS rats and promoted M2 polarization of macrophages. Additionally, inhibition of FoxO1 promoted the proliferation of GCs and inhibited the inflammatory response in GCs. Furthermore, FoxO1 knockdown inhibited the activation of the NF-κB pathway and the formation of the NLRP3 inflammasome in GCs. In conclusion, inhibition of FoxO1 can alleviate PCOS by inhibiting the TLR4/NF-κB/NLRP3 pathway to reduce inflammation and the immune response.


Subject(s)
Forkhead Box Protein O1 , Polycystic Ovary Syndrome , Animals , Female , Rats , Immunity , NF-kappa B , NLR Family, Pyrin Domain-Containing 3 Protein , Polycystic Ovary Syndrome/genetics , Toll-Like Receptor 4 , Forkhead Box Protein O1/genetics , Gene Knockdown Techniques
SELECTION OF CITATIONS
SEARCH DETAIL