Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 343
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 22(8): 947-957, 2021 08.
Article in English | MEDLINE | ID: mdl-34239121

ABSTRACT

One of most challenging issues in tumor immunology is a better understanding of the dynamics in the accumulation of myeloid-derived suppressor cells (MDSCs) in the tumor microenvironment (TIME), as this would lead to the development of new cancer therapeutics. Here, we show that translationally controlled tumor protein (TCTP) released by dying tumor cells is an immunomodulator crucial to full-blown MDSC accumulation in the TIME. We provide evidence that extracellular TCTP mediates recruitment of the polymorphonuclear MDSC (PMN-MDSC) population in the TIME via activation of Toll-like receptor-2. As further proof of principle, we show that inhibition of TCTP suppresses PMN-MDSC accumulation and tumor growth. In human cancers, we find an elevation of TCTP and an inverse correlation of TCTP gene dosage with antitumor immune signatures and clinical prognosis. This study reveals the hitherto poorly understood mechanism of the MDSC dynamics in the TIME, offering a new rationale for cancer immunotherapy.


Subject(s)
Biomarkers, Tumor/metabolism , Chemokine CXCL1/metabolism , Colorectal Neoplasms/immunology , Myeloid-Derived Suppressor Cells/immunology , Toll-Like Receptor 2/immunology , Tumor Microenvironment/immunology , Alarmins/genetics , Alarmins/metabolism , Animals , Biomarkers, Tumor/genetics , Cell Line, Tumor , Female , HEK293 Cells , Humans , Immunotherapy , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , RAW 264.7 Cells , Tumor Protein, Translationally-Controlled 1
2.
Nat Immunol ; 21(8): 892-901, 2020 08.
Article in English | MEDLINE | ID: mdl-32601470

ABSTRACT

Autoreactive T cells are eliminated in the thymus to prevent autoimmunity by promiscuous expression of tissue-restricted self-antigens in medullary thymic epithelial cells. This expression is dependent on the transcription factor Fezf2, as well as the transcriptional regulator Aire, but the entire picture of the transcriptional program has been obscure. Here, we found that the chromatin remodeler Chd4, also called Mi-2ß, plays a key role in the self-antigen expression in medullary thymic epithelial cells. To maximize the diversity of self-antigen expression, Fezf2 and Aire utilized completely distinct transcriptional mechanisms, both of which were under the control of Chd4. Chd4 organized the promoter regions of Fezf2-dependent genes, while contributing to the Aire-mediated induction of self-antigens via super-enhancers. Mice deficient in Chd4 specifically in thymic epithelial cells exhibited autoimmune phenotypes, including T cell infiltration. Thus, Chd4 plays a critical role in integrating Fezf2- and Aire-mediated gene induction to establish central immune tolerance.


Subject(s)
Autoantigens/immunology , Central Tolerance/physiology , Gene Expression Regulation/immunology , Mi-2 Nucleosome Remodeling and Deacetylase Complex/immunology , Animals , Autoantigens/biosynthesis , DNA Helicases/immunology , DNA Helicases/metabolism , HEK293 Cells , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Mice , Mice, Inbred C57BL , Transcription Factors/immunology , Transcription Factors/metabolism , AIRE Protein
3.
Cell ; 163(4): 975-87, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26544942

ABSTRACT

Self-tolerance to immune reactions is established via promiscuous expression of tissue-restricted antigens (TRAs) in medullary thymic epithelial cells (mTECs), leading to the elimination of T cells that respond to self-antigens. The transcriptional regulator Aire has been thought to be sufficient for the induction of TRAs, despite some indications that other factors may promote TRA expression in the thymus. Here, we show that the transcription factor Fezf2 directly regulates various TRA genes in mTECs independently of Aire. Mice lacking Fezf2 in mTECs displayed severe autoimmune symptoms, including the production of autoantibodies and inflammatory cell infiltration targeted to peripheral organs. These responses differed from those detected in Aire-deficient mice. Furthermore, Fezf2 expression and Aire expression are regulated by distinct signaling pathways and promote the expression of different classes of proteins. Thus, two independent factors, Fezf2 and Aire, permit the expression of TRAs in the thymus to ensure immune tolerance.


Subject(s)
DNA-Binding Proteins/metabolism , Immune Tolerance , Nerve Tissue Proteins/metabolism , Thymus Gland/immunology , Animals , Autoantigens/immunology , Autoimmunity , DNA-Binding Proteins/genetics , Humans , Mice , Nerve Tissue Proteins/genetics , Signal Transduction , Thymocytes/immunology , Transcription Factors/metabolism , AIRE Protein
4.
EMBO J ; 42(22): e114032, 2023 Nov 15.
Article in English | MEDLINE | ID: mdl-37781951

ABSTRACT

Bone marrow-derived cells (BMDCs) infiltrate hypoxic tumors at a pre-angiogenic state and differentiate into mature macrophages, thereby inducing pro-tumorigenic immunity. A critical factor regulating this differentiation is activation of SREBP2-a well-known transcription factor participating in tumorigenesis progression-through unknown cellular mechanisms. Here, we show that hypoxia-induced Golgi disassembly and Golgi-ER fusion in monocytic myeloid cells result in nuclear translocation and activation of SREBP2 in a SCAP-independent manner. Notably, hypoxia-induced SREBP2 activation was only observed in an immature lineage of bone marrow-derived cells. Single-cell RNA-seq analysis revealed that SREBP2-mediated cholesterol biosynthesis was upregulated in HSCs and monocytes but not in macrophages in the hypoxic bone marrow niche. Moreover, inhibition of cholesterol biosynthesis impaired tumor growth through suppression of pro-tumorigenic immunity and angiogenesis. Thus, our findings indicate that Golgi-ER fusion regulates SREBP2-mediated metabolic alteration in lineage-specific BMDCs under hypoxia for tumor progression.


Subject(s)
Monocytes , Neoplasms , Humans , Monocytes/metabolism , Bone Marrow , Cholesterol/metabolism , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/metabolism , Hypoxia
5.
N Engl J Med ; 387(21): 1923-1934, 2022 Nov 24.
Article in English | MEDLINE | ID: mdl-36342113

ABSTRACT

BACKGROUND: High triglyceride levels are associated with increased cardiovascular risk, but whether reductions in these levels would lower the incidence of cardiovascular events is uncertain. Pemafibrate, a selective peroxisome proliferator-activated receptor α modulator, reduces triglyceride levels and improves other lipid levels. METHODS: In a multinational, double-blind, randomized, controlled trial, we assigned patients with type 2 diabetes, mild-to-moderate hypertriglyceridemia (triglyceride level, 200 to 499 mg per deciliter), and high-density lipoprotein (HDL) cholesterol levels of 40 mg per deciliter or lower to receive pemafibrate (0.2-mg tablets twice daily) or matching placebo. Eligible patients were receiving guideline-directed lipid-lowering therapy or could not receive statin therapy without adverse effects and had low-density lipoprotein (LDL) cholesterol levels of 100 mg per deciliter or lower. The primary efficacy end point was a composite of nonfatal myocardial infarction, ischemic stroke, coronary revascularization, or death from cardiovascular causes. RESULTS: Among 10,497 patients (66.9% with previous cardiovascular disease), the median baseline fasting triglyceride level was 271 mg per deciliter, HDL cholesterol level 33 mg per deciliter, and LDL cholesterol level 78 mg per deciliter. The median follow-up was 3.4 years. As compared with placebo, the effects of pemafibrate on lipid levels at 4 months were -26.2% for triglycerides, -25.8% for very-low-density lipoprotein (VLDL) cholesterol, -25.6% for remnant cholesterol (cholesterol transported in triglyceride-rich lipoproteins after lipolysis and lipoprotein remodeling), -27.6% for apolipoprotein C-III, and 4.8% for apolipoprotein B. A primary end-point event occurred in 572 patients in the pemafibrate group and in 560 of those in the placebo group (hazard ratio, 1.03; 95% confidence interval, 0.91 to 1.15), with no apparent effect modification in any prespecified subgroup. The overall incidence of serious adverse events did not differ significantly between the groups, but pemafibrate was associated with a higher incidence of adverse renal events and venous thromboembolism and a lower incidence of nonalcoholic fatty liver disease. CONCLUSIONS: Among patients with type 2 diabetes, mild-to-moderate hypertriglyceridemia, and low HDL and LDL cholesterol levels, the incidence of cardiovascular events was not lower among those who received pemafibrate than among those who received placebo, although pemafibrate lowered triglyceride, VLDL cholesterol, remnant cholesterol, and apolipoprotein C-III levels. (Funded by the Kowa Research Institute; PROMINENT ClinicalTrials.gov number, NCT03071692.).


Subject(s)
Cardiovascular Diseases , Diabetes Mellitus, Type 2 , Hypertriglyceridemia , Hypolipidemic Agents , PPAR alpha , Humans , Apolipoprotein C-III/blood , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/etiology , Cholesterol/blood , Cholesterol, LDL/blood , Diabetes Mellitus, Type 2/complications , Double-Blind Method , Heart Disease Risk Factors , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hyperlipidemias/blood , Hyperlipidemias/drug therapy , Hypertriglyceridemia/blood , Hypertriglyceridemia/complications , Hypertriglyceridemia/drug therapy , Risk Factors , Triglycerides/blood , Hypolipidemic Agents/therapeutic use , PPAR alpha/agonists , Cholesterol, HDL/blood
6.
EMBO J ; 39(20): e105130, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32914505

ABSTRACT

Silkworm ovarian germ cells produce the Siwi-piRNA-induced silencing complex (piRISC) through two consecutive mechanisms, the primary pathway and the secondary ping-pong cycle. Primary Siwi-piRISC production occurs on the outer mitochondrial membrane in an Ago3-independent manner, where Tudor domain-containing Papi binds unloaded Siwi via its symmetrical dimethylarginines (sDMAs). Here, we now show that secondary Siwi-piRISC production occurs at the Ago3-positive nuage Ago3 bodies, in an Ago3-dependent manner, where Vreteno (Vret), another Tudor protein, interconnects unloaded Siwi and Ago3-piRISC through their sDMAs. Upon Siwi depletion, Ago3 is phosphorylated and insolubilized in its piRISC form with cleaved RNAs and Vret, suggesting that the complex is stalled in the intermediate state. The Ago3 bodies are also enlarged. The aberrant morphology is restored upon Siwi re-expression without Ago3-piRISC supply. Thus, Siwi depletion aggregates the Ago3 bodies to protect the piRNA intermediates from degradation until the normal cellular environment returns to re-initiate the ping-pong cycle. Overall, these findings reveal a unique regulatory mechanism controlling piRNA biogenesis.


Subject(s)
Argonaute Proteins/metabolism , Bombyx/metabolism , Germ Cells/metabolism , Insect Proteins/metabolism , RNA, Small Interfering/metabolism , Tudor Domain/genetics , Animals , Arginine/analogs & derivatives , Arginine/metabolism , Argonaute Proteins/genetics , Bombyx/genetics , Bombyx/growth & development , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cells, Cultured , Chromatography, Liquid , Computational Biology , Female , Insect Proteins/genetics , Ovary/cytology , Ovary/metabolism , Phosphorylation , RNA Interference , RNA, Small Interfering/genetics , RNA-Seq , Tandem Mass Spectrometry
7.
Bioorg Med Chem Lett ; 108: 129803, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38777280

ABSTRACT

Targeted delivery of radionuclides to tumors is significant in theranostics applications for precision medicine. Pre-targeting, in which a tumor-targeting vehicle and a radionuclide-loaded effector small molecule are administered separately, holds promise since it can reduce unnecessary internal radiation exposure of healthy cells and can minimize radiation decay. The success of the pre-targeting delivery requires an in vivo-stable tumor-targeting vehicle selectively binding to tumor antigens and an in vivo-stable small molecule effector selectively binding to the vehicle accumulated on the tumor. We previously reported a drug delivery system composed of a low-immunogenic streptavidin with weakened affinity to endogenous biotin and a bis-iminobiotin with high affinity to the engineered streptavidin. It was, however, unknown whether the bis-iminobiotin is stable in vivo when administered alone for the pre-targeting applications. Here we report a new in vivo-stable bis-iminobiotin derivative. The keys to success were the identification of the degradation site of the original bis-iminobiotin treated with mouse plasma and the structural modification of the degradation site. We disclosed the successful pre-targeting delivery of astatine-211 (211At), α-particle emitter, to the CEACAM5-positive tumor in xenograft mouse models.


Subject(s)
Biotin , Streptavidin , Animals , Streptavidin/chemistry , Mice , Biotin/chemistry , Humans , Drug Delivery Systems , Cell Line, Tumor , Mutation , Molecular Structure
8.
Nature ; 555(7695): 260-264, 2018 03 08.
Article in English | MEDLINE | ID: mdl-29489748

ABSTRACT

PIWI-interacting RNAs (piRNAs) are small regulatory RNAs that bind to PIWI proteins to control transposons and maintain genome integrity in animal germ lines. piRNA 3' end formation in the silkworm Bombyx mori has been shown to be mediated by the 3'-to-5' exonuclease Trimmer (Trim; known as PNLDC1 in mammals), and piRNA intermediates are bound with PIWI anchored onto mitochondrial Tudor domain protein Papi. However, it remains unclear whether the Zucchini (Zuc) endonuclease and Nibbler (Nbr) 3'-to-5' exonuclease, both of which have pivotal roles in piRNA biogenesis in Drosophila, are required for piRNA processing in other species. Here we show that the loss of Zuc in Bombyx had no effect on the levels of Trim and Nbr, but resulted in the aberrant accumulation of piRNA intermediates within the Papi complex, and that these were processed to form mature piRNAs by recombinant Zuc. Papi exerted its RNA-binding activity only when bound with PIWI and phosphorylated, suggesting that complex assembly involves a hierarchical process. Both the 5' and 3' ends of piRNA intermediates within the Papi complex showed hallmarks of PIWI 'slicer' activity, yet no phasing pattern was observed in mature piRNAs. The loss of Zuc did not affect the 5'- and 3'-end formation of the intermediates, strongly supporting the idea that the 5' end of Bombyx piRNA is formed by PIWI slicer activity, but independently of Zuc, whereas the 3' end is formed by the Zuc endonuclease. The Bombyx piRNA biogenesis machinery is simpler than that of Drosophila, because Bombyx has no transcriptional silencing machinery that relies on phased piRNAs.


Subject(s)
Bombyx/cytology , Bombyx/genetics , Endoribonucleases/metabolism , Germ Cells/metabolism , Mitochondrial Proteins/metabolism , RNA, Small Interfering/biosynthesis , Animals , Argonaute Proteins/metabolism , Drosophila , RNA, Small Interfering/genetics
9.
Mol Psychiatry ; 27(3): 1694-1703, 2022 03.
Article in English | MEDLINE | ID: mdl-34997193

ABSTRACT

The amygdala, a critical brain region responsible for emotional behavior, is crucially involved in the regulation of the effects of stress on emotional behavior. In the mammalian forebrain, gastrin-releasing peptide (GRP), a 27-amino-acid mammalian neuropeptide, which is a homolog of the 14-amino-acid amidated amphibian peptide bombesin, is highly expressed in the amygdala. The levels of GRP are markedly increased in the amygdala after acute stress; therefore, it is known as a stress-activated modulator. To determine the role of GRP in emotional behavior under stress, we conducted some behavioral and biochemical experiments with GRP-knockout (KO) mice. GRP-KO mice exhibited a longer freezing response than wild-type (WT) littermates in both contextual and auditory fear (also known as threat) conditioning tests only when they were subjected to acute restraint stress 20 min before the conditioning. To identify the critical neural circuits associated with the regulation of emotional memory by GRP, we conducted Arc/Arg3.1-reporter mapping in the amygdala with an Arc-Venus reporter transgenic mouse line. In the amygdalostriatal transition area (AST) and the lateral side of the basal nuclei, fear conditioning after restraint stress increased neuronal activity significantly in WT mice, and GRP KO was found to negate this potentiation only in the AST. These results indicate that the GRP-activated neurons in the AST are likely to suppress excessive fear expression through the regulation of downstream circuits related to fear learning following acute stress.


Subject(s)
Bombesin , Fear , Amygdala/metabolism , Animals , Bombesin/metabolism , Bombesin/pharmacology , Conditioning, Classical/physiology , Fear/physiology , Gastrin-Releasing Peptide/metabolism , Gastrin-Releasing Peptide/pharmacology , Mammals/metabolism , Mice , Mice, Knockout
10.
Cell ; 132(5): 794-806, 2008 Mar 07.
Article in English | MEDLINE | ID: mdl-18329366

ABSTRACT

Certain autoimmune diseases result in abnormal bone homeostasis, but association of immunodeficiency with bone is poorly understood. Osteoclasts, which derive from bone marrow cells, are under the control of the immune system. Differentiation of osteoclasts is mainly regulated by signaling pathways activated by RANK and immune receptors linked to ITAM-harboring adaptors. However, it is unclear how the two signals merge to cooperate in osteoclast differentiation. Here we report that mice lacking the tyrosine kinases Btk and Tec show severe osteopetrosis caused by a defect in bone resorption. RANK and ITAM signaling results in formation of a Btk(Tec)/BLNK(SLP-76)-containing complex and PLCgamma-mediated activation of an essential calcium signal. Furthermore, Tec kinase inhibition reduces osteoclastic bone resorption in models of osteoporosis and inflammation-induced bone destruction. Thus, this study reveals the importance of the osteoclastogenic signaling complex composed of tyrosine kinases, which may provide the molecular basis for a new therapeutic strategy.


Subject(s)
Cell Differentiation , Osteoclasts/cytology , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Agammaglobulinaemia Tyrosine Kinase , Amino Acid Motifs , Animals , Bone and Bones/cytology , Bone and Bones/metabolism , Calcium Signaling , Disease Models, Animal , Female , Mice , Osteoclasts/metabolism , Osteopetrosis/drug therapy , Osteopetrosis/genetics , Osteopetrosis/metabolism , Osteoporosis/metabolism , Osteoporosis/pathology , Phospholipase C gamma/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , RANK Ligand/metabolism , RANK Ligand/pharmacology
11.
Mol Cell ; 60(4): 584-96, 2015 Nov 19.
Article in English | MEDLINE | ID: mdl-26590716

ABSTRACT

Bivalent H3K4me3 and H3K27me3 chromatin domains in embryonic stem cells keep active developmental regulatory genes expressed at very low levels and poised for activation. Here, we show an alternative and previously unknown bivalent modified histone signature in lineage-committed mesenchymal stem cells and preadipocytes that pairs H3K4me3 with H3K9me3 to maintain adipogenic master regulatory genes (Cebpa and Pparg) expressed at low levels yet poised for activation when differentiation is required. We show lineage-specific gene-body DNA methylation recruits H3K9 methyltransferase SETDB1, which methylates H3K9 immediately downstream of transcription start sites marked with H3K4me3 to establish the bivalent domain. At the Cebpa locus, this prevents transcription factor C/EBPß binding, histone acetylation, and further H3K4me3 deposition and is associated with pausing of RNA polymerase II, which limits Cebpa gene expression and adipogenesis.


Subject(s)
Adipocytes/cytology , CCAAT-Enhancer-Binding Proteins/metabolism , DNA Methylation , Histones/genetics , PPAR gamma/metabolism , 3T3 Cells , Adipocytes/physiology , Animals , Cell Differentiation , Cell Lineage , Cells, Cultured , Chromatin/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Histones/chemistry , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Mice , Protein Structure, Tertiary
12.
J Biol Chem ; 296: 100697, 2021.
Article in English | MEDLINE | ID: mdl-33895138

ABSTRACT

Down syndrome critical region (DSCR)-1 functions as a feedback modulator for calcineurin-nuclear factor for activated T cell (NFAT) signals, which are crucial for cell proliferation and inflammation. Stable expression of DSCR-1 inhibits pathological angiogenesis and septic inflammation. DSCR-1 also plays a critical role in vascular wall remodeling associated with aneurysm development that occurs primarily in smooth muscle cells. Besides, Dscr-1 deficiency promotes the M1-to M2-like phenotypic switch in macrophages, which correlates to the reduction of denatured cholesterol uptakes. However, the distinct roles of DSCR-1 in cholesterol and lipid metabolism are not well understood. Here, we show that loss of apolipoprotein (Apo) E in mice with chronic hypercholesterolemia induced Dscr-1 expression in the liver and aortic atheroma. In Dscr-1-null mice fed a high-fat diet, oxidative- and endoplasmic reticulum (ER) stress was induced, and sterol regulatory element-binding protein (SREBP) 2 production in hepatocytes was stimulated. This exaggerated ApoE-/--mediated nonalcoholic fatty liver disease (NAFLD) and subsequent hypercholesterolemia. Genome-wide screening revealed that loss of both ApoE and Dscr-1 resulted in the induction of immune- and leukocyte activation-related genes in the liver compared with ApoE deficiency alone. However, expressions of inflammation-activated markers and levels of monocyte adhesion were suspended upon induction of the Dscr-1 null background in the aortic endothelium. Collectively, our study shows that the combined loss of Dscr-1 and ApoE causes metabolic dysfunction in the liver but reduces atherosclerotic plaques, thereby leading to a dramatic increase in serum cholesterol and the formation of sporadic vasculopathy.


Subject(s)
Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Calcium-Binding Proteins/deficiency , Cholesterol/metabolism , Gene Deletion , Hypercholesterolemia/genetics , Muscle Proteins/deficiency , Animals , Calcium-Binding Proteins/genetics , Gene Expression Regulation , Hepatocytes/metabolism , Hypercholesterolemia/metabolism , Mice , Muscle Proteins/genetics , Phenotype
13.
Cancer Sci ; 113(12): 4350-4362, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36121618

ABSTRACT

Antibody-mimetic drug conjugate is a novel noncovalent conjugate consisting of an antibody-mimetic recognizing a target molecule on the cancer cell surface and low-molecular-weight payloads that kill the cancer cells. In this study, the efficacy of a photo-activating antibody-mimetic drug conjugate targeting HER2-expressing tumors was evaluated in mice, by using the affibody that recognize HER2 (ZHER2:342 ) as a target molecule and an axially substituted silicon phthalocyanine (a novel potent photo-activating compound) as a payload. The first treatment with the photo-activating antibody-mimetic drug conjugates reduced the size of all HER2-expressing KPL-4 xenograft tumors macroscopically. However, during the observation period, relapsed tumors gradually appeared in approximately 50% of the animals. To evaluate the efficacy of repeated antibody-mimetic drug conjugate treatment, animals with relapsed tumors were treated again with the same regimen. After the second observation period, the mouse tissues were examined histopathologically. Unexpectedly, all relapsed tumors were eradicated, and all animals were diagnosed with pathological complete remission. After the second treatment, skin wounds healed rapidly, and no significant side effects were observed in other organs, except for occasional microscopic granulomatous tissues beneath the serosa of the liver in a few mice. Repeated treatments seemed to be well tolerated. These results indicate the promising efficacy of the repeated photo-activating antibody-mimetic drug conjugate treatment against HER2-expressing tumors.


Subject(s)
Immunoconjugates , Humans , Animals , Mice , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Receptor, ErbB-2/metabolism , Cell Line, Tumor , Antibodies
14.
Genes Cells ; 26(7): 513-529, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33971063

ABSTRACT

The lysine methyltransferase SETDB1, an enzyme responsible for methylation of histone H3 at lysine 9, plays a key role in H3K9 tri-methylation-dependent silencing of endogenous retroviruses and developmental genes. Recent studies have shown that ubiquitination of human SETDB1 complements its catalytic activity and the silencing of endogenous retroviruses in human embryonic stem cells. However, it is not known whether SETDB1 ubiquitination is essential for its other major role in epigenetic silencing of developmental gene programs. We previously showed that SETDB1 contributes to the formation of H3K4/H3K9me3 bivalent chromatin domains that keep adipogenic Cebpa and Pparg genes in a poised state for activation and restricts the differentiation potential of pre-adipocytes. Here, we show that ubiquitin-resistant K885A mutant of SETDB1 represses adipogenic genes and inhibits pre-adipocyte differentiation similar to wild-type SETDB1. We show this was due to a compensation mechanism for H3K9me3 chromatin modifications on the Cebpa locus by other H3K9 methyltransferases Suv39H1 and Suv39H2. In contrast, the K885A mutant did not repress other SETDB1 target genes such as Tril and Gas6 suggesting SETDB1 represses its target genes by two mechanisms; one that requires its ubiquitination and another that still requires SETDB1 but not its enzyme activity.


Subject(s)
Adipogenesis , Epigenesis, Genetic , Histone-Lysine N-Methyltransferase/metabolism , Ubiquitination , 3T3-L1 Cells , Animals , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , HEK293 Cells , Histone Code , Histone-Lysine N-Methyltransferase/genetics , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mutation, Missense
15.
Protein Expr Purif ; 192: 106043, 2022 04.
Article in English | MEDLINE | ID: mdl-34973460

ABSTRACT

Antibody-drug conjugates (ADCs) are a major therapeutic tool for the treatment of advanced cancer. Malignant cells in advanced cancer often display multiple genetic mutations and become resistant to monotherapy. Therefore, a therapeutic regimen that simultaneously targets multiple molecules with multiple payloads is desirable. However, the development of ADCs is hampered by issues in biopharmaceutical manufacturing and the complexity of the conjugation process of low-molecular-weight payloads to biologicals. Here, we report antibody mimetic-drug conjugates (AMDCs) developed by exploiting the non-covalent binding property of payloads based on high-affinity binding of mutated streptavidin and modified iminobiotin. Miniprotein antibodies were fused to a low immunogenic streptavidin variant, which was then expressed in Escherichia coli inclusion bodies, solubilized, and refolded into functional tetramers. The AMDC developed against human epidermal growth factor receptor 2 (HER2) effectively killed cultured cancer cells using bis-iminobiotin conjugated to photo-activating silicon phthalocyanine. The HER2-targeting AMDC was also effective in vivo against a mouse KPL-4 xenograft model. This AMDC platform provides rapid, stable, and high-yield therapeutics against multiple targets.


Subject(s)
Escherichia coli/metabolism , Gene Expression , Immunoconjugates/genetics , Animals , Biotin/administration & dosage , Biotin/analogs & derivatives , Biotin/chemistry , Biotin/genetics , Biotin/immunology , Cell Line, Tumor , Cloning, Molecular , Escherichia coli/genetics , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/chemistry , Immunoconjugates/immunology , Mice , Mice, Inbred BALB C , Neoplasms/drug therapy , Protein Folding , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , Streptavidin/administration & dosage , Streptavidin/chemistry , Streptavidin/genetics , Streptavidin/immunology
16.
J Epidemiol ; 32(2): 105-111, 2022 02 05.
Article in English | MEDLINE | ID: mdl-34776499

ABSTRACT

BACKGROUND: Tokyo, the capital of Japan, is a densely populated city of >13 million people, so the population is at high risk of epidemic severe acute respiratory coronavirus 2 (SARS-CoV-2) infection. A serologic survey of anti-SARS-CoV-2 IgG would provide valuable data for assessing the city's SARS-CoV-2 infection status. Therefore, this cross-sectional study estimated the anti-SARS-CoV-2 IgG seroprevalence in Tokyo. METHODS: Leftover serum of 23,234 hospital visitors was tested for antibodies against SARS-CoV-2 using an iFlash 3000 chemiluminescence immunoassay analyzer (Shenzhen YHLO Biotech, Shenzhen, China) with an iFlash-SARS-CoV-2 IgG kit (YHLO) and iFlash-SARS-CoV-2 IgG-S1 kit (YHLO). Serum samples with a positive result (≥10 AU/mL) in either of these assays were considered seropositive for anti-SARS-CoV-2 IgG. Participants were randomly selected from patients visiting 14 Tokyo hospitals between September 1, 2020 and March 31, 2021. No participants were diagnosed with coronavirus disease 2019 (COVID-19), and none exhibited COVID-19-related symptoms at the time of blood collection. RESULTS: The overall anti-SARS-CoV-2 IgG seroprevalence among all participants was 1.83% (95% confidence interval [CI], 1.66-2.01%). The seroprevalence in March 2021, the most recent month of this study, was 2.70% (95% CI, 2.16-3.34%). After adjusting for population age, sex, and region, the estimated seroprevalence in Tokyo was 3.40%, indicating that 470,778 individuals had a history of SARS-CoV-2 infection. CONCLUSIONS: The estimated number of individuals in Tokyo with a history of SARS-CoV-2 infection was 3.9-fold higher than the number of confirmed cases. Our study enhances understanding of the SARS-CoV-2 epidemic in Tokyo.


Subject(s)
COVID-19 , Antibodies, Viral , Cross-Sectional Studies , Hospitals , Humans , Immunoglobulin G , SARS-CoV-2 , Seroepidemiologic Studies , Tokyo/epidemiology
17.
Mol Cell ; 53(3): 393-406, 2014 Feb 06.
Article in English | MEDLINE | ID: mdl-24507715

ABSTRACT

Although thousands of long noncoding RNAs (lncRNAs) are localized in the nucleus, only a few dozen have been functionally characterized. Here we show that nuclear enriched abundant transcript 1 (NEAT1), an essential lncRNA for the formation of nuclear body paraspeckles, is induced by influenza virus and herpes simplex virus infection as well as by Toll-like receptor3-p38 pathway-triggered poly I:C stimulation, resulting in excess formation of paraspeckles. We found that NEAT1 facilitates the expression of antiviral genes including cytokines such as interleukin-8 (IL8). We found that splicing factor proline/glutamine-rich (SFPQ), a NEAT1-binding paraspeckle protein, is a repressor of IL8 transcription, and that NEAT1 induction relocates SFPQ from the IL8 promoter to the paraspeckles, leading to transcriptional activation of IL8. Together, our data show that NEAT1 plays an important role in the innate immune response through the transcriptional regulation of antiviral genes by the stimulus-responsive cooperative action of NEAT1 and SFPQ.


Subject(s)
Immunity, Innate/genetics , Interleukin-8/genetics , RNA, Long Noncoding/physiology , RNA-Binding Proteins/metabolism , Gene Expression Regulation , HeLa Cells , Herpesvirus 1, Human/immunology , Humans , Measles virus/immunology , Orthomyxoviridae/immunology , PTB-Associated Splicing Factor , Promoter Regions, Genetic , Protein Transport , RNA, Long Noncoding/genetics , Transcription, Genetic
18.
Curr Atheroscler Rep ; 23(1): 3, 2021 01 03.
Article in English | MEDLINE | ID: mdl-33392801

ABSTRACT

PURPOSE OF REVIEW: Adoption of poor lifestyles (inactivity and energy-dense diets) has driven the worldwide increase in the metabolic syndrome, type 2 diabetes mellitus and non-alcoholic steatohepatitis (NASH). Of the defining features of the metabolic syndrome, an atherogenic dyslipidaemia characterised by elevated triglycerides (TG) and low plasma concentration of high-density lipoprotein cholesterol is a major driver of risk for atherosclerotic cardiovascular disease. Beyond lifestyle intervention and statins, targeting the nuclear receptor peroxisome proliferator-activated receptor alpha (PPARα) is a therapeutic option. However, current PPARα agonists (fibrates) have limitations, including safety issues and the lack of definitive evidence for cardiovascular benefit. Modulating the ligand structure to enhance binding at the PPARα receptor, with the aim of maximising beneficial effects and minimising adverse effects, underlies the SPPARMα concept. RECENT FINDINGS: This review discusses the history of SPPARM development, latterly focusing on evidence for the first licensed SPPARMα, pemafibrate. Evidence from animal models of hypertriglyceridaemia or NASH, as well as clinical trials in patients with atherogenic dyslipidaemia, are overviewed. The available data set the scene for therapeutic application of SPPARMα in the metabolic syndrome, and possibly, NASH. The outstanding question, which has so far eluded fibrates in the setting of current evidence-based therapy including statins, is whether treatment with pemafibrate significantly reduces cardiovascular events in patients with atherogenic dyslipidaemia. The PROMINENT study in patients with type 2 diabetes mellitus and this dyslipidaemia is critical to evaluating this.


Subject(s)
Benzoxazoles/therapeutic use , Butyrates/therapeutic use , Metabolic Syndrome/drug therapy , Metabolic Syndrome/metabolism , PPAR alpha/metabolism , Animals , Humans
19.
Biochem Biophys Res Commun ; 522(4): 931-938, 2020 02 19.
Article in English | MEDLINE | ID: mdl-31806376

ABSTRACT

FLCN is a tumor suppressor gene which controls energy homeostasis through regulation of a variety of metabolic pathways including mitochondrial oxidative metabolism and autophagy. Birt-Hogg-Dubé (BHD) syndrome which is driven by germline alteration of the FLCN gene, predisposes patients to develop kidney cancer, cutaneous fibrofolliculomas, pulmonary cysts and less frequently, salivary gland tumors. Here, we report metabolic roles for FLCN in the salivary gland as well as their clinical relevance. Screening of salivary glands of BHD patients using ultrasonography demonstrated increased cyst formation in the salivary gland. Salivary gland tumors that developed in BHD patients exhibited an upregulated mTOR-S6R pathway as well as increased GPNMB expression, which are characteristics of FLCN-deficient cells. Salivary gland-targeted Flcn knockout mice developed cytoplasmic clear cell formation in ductal cells with increased mitochondrial biogenesis, upregulated mTOR-S6K pathway, upregulated TFE3-GPNMB axis and upregulated lipid metabolism. Proteomic and metabolite analysis using LC/MS and GC/MS revealed that Flcn inactivation in salivary gland triggers metabolic reprogramming towards the pentose phosphate pathway which consequently upregulates nucleotide synthesis and redox regulation, further supporting that Flcn controls metabolic homeostasis in salivary gland. These data uncover important roles for FLCN in salivary gland; metabolic reprogramming under FLCN deficiency might increase nucleotide production which may feed FLCN-deficient salivary gland cells to trigger tumor initiation and progression, providing mechanistic insight into salivary gland tumorigenesis as well as a foundation for development of novel therapeutics for salivary gland tumors.


Subject(s)
Cysts/metabolism , Cysts/pathology , Nucleotides/biosynthesis , Proto-Oncogene Proteins/metabolism , Salivary Glands/metabolism , Salivary Glands/pathology , Tumor Suppressor Proteins/metabolism , Adult , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cysts/diagnostic imaging , Female , Gene Ontology , Glycolysis , Humans , Male , Mice, Knockout , Middle Aged , Organelle Biogenesis , Pentose Phosphate Pathway , Proto-Oncogene Proteins/deficiency , Salivary Glands/diagnostic imaging , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/deficiency , Up-Regulation
20.
J Biol Chem ; 293(26): 10333-10343, 2018 06 29.
Article in English | MEDLINE | ID: mdl-29764933

ABSTRACT

Peroxisome proliferator-activated receptor α (PPARα) is a ligand-activated transcription factor that belongs to the superfamily of nuclear hormone receptors. PPARα is mainly expressed in the liver, where it activates fatty acid oxidation and lipoprotein metabolism and improves plasma lipid profiles. Therefore, PPARα activators are often used to treat patients with dyslipidemia. To discover additional PPARα activators as potential compounds for use in hypolipidemic drugs, here we established human hepatoblastoma cell lines with luciferase reporter expression from the promoters containing peroxisome proliferator-responsive elements (PPREs) and tetracycline-regulated expression of full-length human PPARα to quantify the effects of chemical ligands on PPARα activity. Using the established cell-based PPARα-activator screening system to screen a library of >12,000 chemical compounds, we identified several hit compounds with basic chemical skeletons different from those of known PPARα agonists. One of the hit compounds, a 1H-pyrazolo[3,4-b]pyridine-4-carboxylic acid derivative we termed compound 3, selectively up-regulated PPARα transcriptional activity, leading to PPARα target gene expression both in vitro and in vivo Of note, the half-maximal effective concentrations of the hit compounds were lower than that of the known PPARα ligand fenofibrate. Finally, fenofibrate or compound 3 treatment of high fructose-fed rats having elevated plasma triglyceride levels for 14 days indicated that compound 3 reduces plasma triglyceride levels with similar efficiency as fenofibrate. These observations raise the possibility that 1H-pyrazolo[3,4-b]pyridine-4-carboxylic acid derivatives might be effective drug candidates for selective targeting of PPARα to manage dyslipidemia.


Subject(s)
Gene Expression Regulation , PPAR alpha/genetics , PPAR alpha/metabolism , Animals , Drug Evaluation, Preclinical , Fructose/adverse effects , Gene Expression Regulation/drug effects , Genes, Reporter/genetics , Humans , Hypolipidemic Agents/pharmacology , Ligands , Rats
SELECTION OF CITATIONS
SEARCH DETAIL