Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
Add more filters

Publication year range
1.
J Immunol ; 211(12): 1835-1843, 2023 12 15.
Article in English | MEDLINE | ID: mdl-37930129

ABSTRACT

Oxidative stress induces a prothrombotic state through enhancement of adhesion properties of the endothelium. E-selectin, an endothelial cell adhesion molecule, becomes a therapeutic target for venous thrombosis, whereas the regulatory mechanisms of its expression have not been fully understood. In the present study, we report that H2O2 treatment increases expression of E-selectin but decreases expression of the endothelial transcription factor ETS-related gene (ERG) in HUVECs in a dose- and time-dependent manner. In BALB/c mice treated with hypochlorous acid, E-selectin expression is increased and ERG expression is decreased in endothelial cells of the brain and lung. RNA interference of ERG upregulates E-selectin expression, whereas transfection of ERG-expressing plasmid downregulates E-selectin expression in HUVECs. Knockdown or overexpression of ERG comprises H2O2-induced E-selectin expression in HUVECs. Deletion of the Erg gene in mice results in embryonic lethality at embryonic days 10.5-12.5, and E-selectin expression is increased in the Erg-/- embryos. No chromatin loop was found on the E-selectin gene or its promoter region by capture high-throughput chromosome conformation capture. Chromatin immunoprecipitation and luciferase reporter assay determined that the -127 ERG binding motif mediates ERG-repressed E-selectin promoter activity. In addition, ERG decreases H2O2-induced monocyte adhesion. Together, ERG represses the E-selectin gene transcription and inhibits oxidative stress-induced endothelial cell adhesion.


Subject(s)
E-Selectin , Transcription Factors , Animals , Mice , Transcription Factors/metabolism , E-Selectin/genetics , E-Selectin/metabolism , Endothelial Cells/metabolism , Cells, Cultured , Hydrogen Peroxide/metabolism , Oxidative Stress , Endothelium, Vascular/metabolism
2.
EMBO J ; 39(7): e103949, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32125007

ABSTRACT

Histone H3 lysine-9 di-methylation (H3K9me2) and lysine-27 tri-methylation (H3K27me3) are linked to repression of gene expression, but the functions of repressive histone methylation dynamics during inflammatory responses remain enigmatic. Here, we report that lysine demethylases 7A (KDM7A) and 6A (UTX) play crucial roles in tumor necrosis factor (TNF)-α signaling in endothelial cells (ECs), where they are regulated by a novel TNF-α-responsive microRNA, miR-3679-5p. TNF-α rapidly induces co-occupancy of KDM7A and UTX at nuclear factor kappa-B (NF-κB)-associated elements in human ECs. KDM7A and UTX demethylate H3K9me2 and H3K27me3, respectively, and are both required for activation of NF-κB-dependent inflammatory genes. Chromosome conformation capture-based methods furthermore uncover increased interactions between TNF-α-induced super enhancers at NF-κB-relevant loci, coinciding with KDM7A and UTX recruitments. Simultaneous pharmacological inhibition of KDM7A and UTX significantly reduces leukocyte adhesion in mice, establishing the biological and potential translational relevance of this mechanism. Collectively, these findings suggest that rapid erasure of repressive histone marks by KDM7A and UTX is essential for NF-κB-dependent regulation of genes that control inflammatory responses of ECs.


Subject(s)
Endothelial Cells/immunology , Histone Demethylases/metabolism , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , MicroRNAs/genetics , Animals , Cell Adhesion , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Expression Regulation , Gene Regulatory Networks , Histones/chemistry , Human Umbilical Vein Endothelial Cells , Humans , Lysine/metabolism , Male , Methylation , Mice , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
3.
Dev Dyn ; 2022 Aug 29.
Article in English | MEDLINE | ID: mdl-36038963

ABSTRACT

BACKGROUND: The somatopleure serves as the primordium of the amnion, an extraembryonic membrane surrounding the embryo. Recently, we have reported that amniogenic somatopleural cells (ASCs) not only form the amnion but also migrate into the embryo and differentiate into cardiomyocytes and vascular endothelial cells. However, detailed differentiation processes and final distributions of these intra-embryonic ASCs (hereafter referred to as iASCs) remain largely unknown. RESULTS: By quail-chick chimera analysis, we here show that iASCs differentiate into various cell types including cardiomyocytes, smooth muscle cells, cardiac interstitial cells, and vascular endothelial cells. In the pharyngeal region, they distribute selectively into the thyroid gland and differentiate into vascular endothelial cells to form intra-thyroid vasculature. Explant culture experiments indicated sequential requirement of fibroblast growth factor (FGF) and vascular endothelial growth factor (VEGF) signaling for endothelial differentiation of iASCs. Single-cell transcriptome analysis further revealed heterogeneity and the presence of hemangioblast-like cell population within ASCs, with a switch from FGF to VEGF receptor gene expression. CONCLUSION: The present study demonstrates novel roles of ASCss especially in heart and thyroid development. It will provide a novel clue for understanding the cardiovascular development of amniotes from embryological and evolutionary perspectives.

4.
Biochem Biophys Res Commun ; 590: 89-96, 2022 01 29.
Article in English | MEDLINE | ID: mdl-34973535

ABSTRACT

Cholinergic anti-inflammatory pathway (CAP) describes a neuronal-inflammatory reflex centered on systemic cytokine regulation by α7 nicotinic acetylcholine receptor (α7nAChR) activation of spleen-residue macrophage. However, the CAP mechanism attenuating distal tissue inflammation, inducing a low level of systemic inflammation, is lesser known. In this study, we hypothesized that CAP regulates monocyte accessibility by influencing their adhesion to endothelial cells. Using RNA-seq analysis, we identified that α1,3-Fucosyltransferase 7 (FucT-VII), the enzyme required for processing selectin ligands, was significantly downregulated by α7nAChR agonist among other cell-cell adhesion genes. The α7nAChR agonist inhibited monocytic cell line U-937 binding to P-selectin and adhesion to endothelial cells. Furthermore, α7nAChR agonist selectivity was confirmed by α7nAChR knockdown assays, showing that FUT7 inhibition and adhesion attenuation by the agonist was abolished by siRNA targeting α7nAChR encoding gene. Consistently, FUT7 knockdown inhibited the adhesive properties of U-937 and prevented them to adhere to endothelial cells. Overexpression of FUT7 also abrogated the adhesion attenuation induced by GTS-21 indicating that FUT7 inhibition was sufficient for inhibiting adhesion by α7nAChR activation. Our work demonstrated that α7nAChR activation regulates monocyte adhesion to endothelial cells through FUT7 inhibition, providing a novel insight into the CAP mechanism.


Subject(s)
Fucosyltransferases/antagonists & inhibitors , Human Umbilical Vein Endothelial Cells/cytology , Monocytes/cytology , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Benzylidene Compounds/pharmacology , Cell Adhesion/drug effects , Cell Adhesion/genetics , Down-Regulation/drug effects , Down-Regulation/genetics , Fucosyltransferases/metabolism , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Monocytes/drug effects , Monocytes/metabolism , Pyridines/pharmacology , U937 Cells , alpha7 Nicotinic Acetylcholine Receptor/antagonists & inhibitors
5.
Biochem Biophys Res Commun ; 605: 16-23, 2022 05 21.
Article in English | MEDLINE | ID: mdl-35306360

ABSTRACT

Vascular endothelial growth factor (VEGF) signaling plays a central role in vascular development and maintenance of vascular homeostasis. In endothelial cells (ECs), VEGF activates the gene expression of angiogenic transcription factors (TFs), followed by induction of downstream angiogenic responsive genes. Recent findings support that histone modification dynamics contribute to the transcriptional control of genes that are important for EC functions. Lysine demethylase 2B (KDM2B) demethylates histone H3K4me3 and H3K36me2/3 and mediates the monoubiquitination of histone H2AK119. KDM2B functions as a transcriptional repressor in somatic cell reprogramming and tumor development. However, the role of KDM2B in VEGF signaling remains to be elucidated. Here, we show that KDM2B knockdown enhances VEGF-induced angiogenesis in cultured human ECs via increased migration and proliferation. In contrast, ectopic expression of KDM2B inhibits angiogenesis. The function of KDM2B may depend on its catalytic Jumonji C domain. Genome-wide analysis further reveals that KDM2B selectively controls the transcription of VEGF-induced angiogenic TFs that are associated with increased H3K4me3/H3K36me3 and decreased H2AK119ub. These findings suggest an essential role of KDM2B in VEGF signaling in ECs. As dysregulation of VEGF signaling in ECs is involved in various diseases, including cancer, KDM2B may be a potential therapeutic target in VEGF-mediated vasculopathic diseases.


Subject(s)
F-Box Proteins , Histones , Cell Proliferation , Endothelial Cells/metabolism , F-Box Proteins/genetics , F-Box Proteins/metabolism , Histones/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Lysine/metabolism , Transcription Factors/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
6.
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
7.
J Am Soc Nephrol ; 32(7): 1599-1615, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33875568

ABSTRACT

BACKGROUND: The sympathetic nervous system regulates immune cell dynamics. However, the detailed role of sympathetic signaling in inflammatory diseases is still unclear because it varies according to the disease situation and responsible cell types. This study focused on identifying the functions of sympathetic signaling in macrophages in LPS-induced sepsis and renal ischemia-reperfusion injury (IRI). METHODS: We performed RNA sequencing of mouse macrophage cell lines to identify the critical gene that mediates the anti-inflammatory effect of ß2-adrenergic receptor (Adrb2) signaling. We also examined the effects of salbutamol (a selective Adrb2 agonist) in LPS-induced systemic inflammation and renal IRI. Macrophage-specific Adrb2 conditional knockout (cKO) mice and the adoptive transfer of salbutamol-treated macrophages were used to assess the involvement of macrophage Adrb2 signaling. RESULTS: In vitro, activation of Adrb2 signaling in macrophages induced the expression of T cell Ig and mucin domain 3 (Tim3), which contributes to anti-inflammatory phenotypic alterations. In vivo, salbutamol administration blocked LPS-induced systemic inflammation and protected against renal IRI; this protection was mitigated in macrophage-specific Adrb2 cKO mice. The adoptive transfer of salbutamol-treated macrophages also protected against renal IRI. Single-cell RNA sequencing revealed that this protection was associated with the accumulation of Tim3-expressing macrophages in the renal tissue. CONCLUSIONS: The activation of Adrb2 signaling in macrophages induces anti-inflammatory phenotypic alterations partially via the induction of Tim3 expression, which blocks LPS-induced systemic inflammation and protects against renal IRI.

8.
Int J Mol Sci ; 23(1)2022 Jan 02.
Article in English | MEDLINE | ID: mdl-35008922

ABSTRACT

The RNA exosome is a multi-subunit ribonuclease complex that is evolutionally conserved and the major cellular machinery for the surveillance, processing, degradation, and turnover of diverse RNAs essential for cell viability. Here we performed integrated genomic and clinicopathological analyses of 27 RNA exosome components across 32 tumor types using The Cancer Genome Atlas PanCancer Atlas Studies' datasets. We discovered that the EXOSC4 gene, which encodes a barrel component of the RNA exosome, was amplified across multiple cancer types. We further found that EXOSC4 alteration is associated with a poor prognosis of pancreatic cancer patients. Moreover, we demonstrated that EXOSC4 is required for the survival of pancreatic cancer cells. EXOSC4 also repressed BIK expression and destabilized SESN2 mRNA by promoting its degradation. Furthermore, knockdown of BIK and SESN2 could partially rescue pancreatic cells from the reduction in cell viability caused by EXOSC4 knockdown. Our study provides evidence for EXOSC4-mediated regulation of BIK and SESN2 mRNA in the survival of pancreatic tumor cells.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Exosome Multienzyme Ribonuclease Complex/genetics , Gene Amplification , Mitochondrial Proteins/metabolism , Nuclear Proteins/metabolism , Pancreatic Neoplasms/metabolism , RNA-Binding Proteins/genetics , Apoptosis Regulatory Proteins/genetics , Cell Line, Tumor , Cell Survival , Exosome Multienzyme Ribonuclease Complex/metabolism , Exosomes/metabolism , Gene Expression Regulation, Neoplastic , Humans , Mitochondrial Proteins/genetics , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/physiopathology , Nuclear Proteins/genetics , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/physiopathology , Prognosis , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , RNA-Seq
9.
Biochem Biophys Res Commun ; 527(4): 993-999, 2020 07 05.
Article in English | MEDLINE | ID: mdl-32446559

ABSTRACT

Most viruses inhibit the innate immune system and/or the RNA degradation processes of host cells to construct an advantageous intracellular environment for their survival. Characteristic RNA sequences within RNA virus genomes or RNAs transcribed from DNA virus genomes contribute toward this inhibition. In this study, we developed a method called "Fate-seq" to comprehensively identify the RNA sequences derived from RNA and DNA viruses, contributing RNA stability in the cells. We examined the stabilization activity of 5,924 RNA fragments derived from 26 different viruses (16 RNA viruses and 10 DNA viruses) using next-generation sequencing of these RNAs fused 3' downstream of GFP reporter RNA. With the Fate-seq approach, we detected multiple virus-derived RNA sequences that stabilized GFP reporter RNA, including sequences derived from severe acute respiratory syndrome-related coronavirus (SARS-CoV). Comparative genomic analysis revealed that these RNA sequences and their predicted secondary structures are highly conserved between SARS-CoV and the novel coronavirus, SARS-CoV-2, which is responsible for the global outbreak of the coronavirus-associated disease that emerged in December 2019 (COVID-19). These sequences have the potential to enhance the stability of viral RNA genomes, thereby augmenting viral replication efficiency and virulence.


Subject(s)
Betacoronavirus/genetics , Coronavirus Infections/virology , Pneumonia, Viral/virology , RNA Stability , RNA, Viral/chemistry , Severe acute respiratory syndrome-related coronavirus/genetics , Base Sequence , Betacoronavirus/chemistry , COVID-19 , Conserved Sequence , Coronaviridae/genetics , Genome, Viral , HeLa Cells , Humans , Nucleic Acid Conformation , Pandemics , Severe acute respiratory syndrome-related coronavirus/chemistry , SARS-CoV-2 , Sequence Analysis, RNA
10.
J Cell Mol Med ; 23(5): 3762-3766, 2019 05.
Article in English | MEDLINE | ID: mdl-30784178

ABSTRACT

Netrin4 (NTN4) is a chemotropic factor that regulates angiogenesis. We found that endothelial expression of the activated, intracellular domain of Notch1 (NICD1), significantly up-regulated NTN4 mRNA as well as intracellular NTN4 protein in both transgenic mice and cultured human umbilical vein endothelial cells (HUVECs). Notch1 activation also increased NTN4 secretion from HUVECs. We subsequently demonstrated that NICD1 bound to CSL (CBF1, Suppressor of Hairless, Lag-1), a core component of Notch transcription complex, at the -53 element of the human NTN4 gene promoter. Loss of the -53 element compromised NICD1-induced NTN4 expression. Our results suggest a conserved role for Notch signalling in transcriptional regulation of endothelial NTN4.


Subject(s)
Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/metabolism , Neovascularization, Physiologic/genetics , Netrins/genetics , Receptor, Notch1/genetics , Animals , Base Sequence , Binding Sites/genetics , Cells, Cultured , Humans , Mice, Transgenic , Netrins/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , Receptor, Notch1/metabolism , Regulatory Elements, Transcriptional/genetics
11.
Kidney Int ; 95(3): 563-576, 2019 03.
Article in English | MEDLINE | ID: mdl-30670317

ABSTRACT

The cholinergic anti-inflammatory pathway (CAP) links the nervous and immune systems and modulates innate and adaptive immunity. Activation of the CAP by vagus nerve stimulation exerts protective effects in a wide variety of clinical disorders including rheumatoid arthritis and Crohn's disease, and in murine models of acute kidney injury including ischemia/reperfusion injury (IRI). The canonical CAP pathway involves activation of splenic alpha7-nicotinic acetylcholine receptor (α7nAChR)-positive macrophages by splenic ß2-adrenergic receptor-positive CD4+ T cells. Here we demonstrate that ultrasound or vagus nerve stimulation also activated α7nAChR-positive peritoneal macrophages, and that adoptive transfer of these activated peritoneal macrophages reduced IRI in recipient mice. The protective effect required α7nAChR, and did not occur in splenectomized mice or in mice lacking T and B cells, suggesting a bidirectional interaction between α7nAChR-positive peritoneal macrophages and other immune cells including ß2-adrenergic receptor-positive CD4+ T cells. We also found that expression of hairy and enhancer of split-1 (Hes1), a basic helix-loop-helix DNA-binding protein, is induced in peritoneal macrophages by ultrasound or vagus nerve stimulation. Adoptive transfer of Hes1-overexpressing peritoneal macrophages reduced kidney IRI. Our data suggest that Hes1 is downstream of α7nAChR and is important to fully activate the CAP. Taken together, these results suggest that peritoneal macrophages play a previously unrecognized role in mediating the protective effect of CAP activation in kidney injury, and that Hes1 is a new candidate pharmacological target to activate the CAP.


Subject(s)
Acute Kidney Injury/immunology , Macrophages, Peritoneal/immunology , Reperfusion Injury/immunology , Transcription Factor HES-1/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/therapy , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/radiation effects , CD4-Positive T-Lymphocytes/transplantation , Disease Models, Animal , Gene Knockdown Techniques , Humans , Macrophage Activation , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/transplantation , Male , Mice , Neuroimmunomodulation/radiation effects , RAW 264.7 Cells , Reperfusion Injury/pathology , Reperfusion Injury/therapy , Transcription Factor HES-1/genetics , Transcription Factor HES-1/immunology , Ultrasonic Therapy , Up-Regulation/radiation effects , Vagus Nerve Stimulation , alpha7 Nicotinic Acetylcholine Receptor/immunology
12.
Nucleic Acids Res ; 45(8): 4344-4358, 2017 05 05.
Article in English | MEDLINE | ID: mdl-28334937

ABSTRACT

Although studies of the differentiation from mouse embryonic stem (ES) cells to vascular endothelial cells (ECs) provide an excellent model for investigating the molecular mechanisms underlying vascular development, temporal dynamics of gene expression and chromatin modifications have not been well studied. Herein, using transcriptomic and epigenomic analyses based on H3K4me3 and H3K27me3 modifications at a genome-wide scale, we analysed the EC differentiation steps from ES cells and crucial epigenetic modifications unique to ECs. We determined that Gata2, Fli1, Sox7 and Sox18 are master regulators of EC that are induced following expression of the haemangioblast commitment pioneer factor, Etv2. These master regulator gene loci were repressed by H3K27me3 throughout the mesoderm period but rapidly transitioned to histone modification switching from H3K27me3 to H3K4me3 after treatment with vascular endothelial growth factor. SiRNA knockdown experiments indicated that these regulators are indispensable not only for proper EC differentiation but also for blocking the commitment to other closely aligned lineages. Collectively, our detailed epigenetic analysis may provide an advanced model for understanding temporal regulation of chromatin signatures and resulting gene expression profiles during EC commitment. These studies may inform the future development of methods to stimulate the vascular endothelium for regenerative medicine.


Subject(s)
Endothelial Cells/metabolism , Epigenesis, Genetic , GATA2 Transcription Factor/genetics , Histones/genetics , Mouse Embryonic Stem Cells/metabolism , Proto-Oncogene Protein c-ets-1/genetics , SOXF Transcription Factors/genetics , Animals , Cell Differentiation , Cell Lineage/genetics , Endothelial Cells/cytology , GATA2 Transcription Factor/antagonists & inhibitors , GATA2 Transcription Factor/metabolism , Histones/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Oligonucleotide Array Sequence Analysis , Primary Cell Culture , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Protein c-ets-1/antagonists & inhibitors , Proto-Oncogene Protein c-ets-1/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , SOXF Transcription Factors/antagonists & inhibitors , SOXF Transcription Factors/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
14.
Dev Biol ; 402(2): 162-74, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25889273

ABSTRACT

Most gnathostomata craniofacial structures derive from pharyngeal arches (PAs), which are colonized by cranial neural crest cells (CNCCs). The anteroposterior and dorsoventral identities of CNCCs are defined by the combinatorial expression of Hox and Dlx genes. The mechanisms associating characteristic Hox/Dlx expression patterns with the topology and morphology of PAs derivatives are only partially known; a better knowledge of these processes might lead to new concepts on the origin of taxon-specific craniofacial morphologies and of certain craniofacial malformations. Here we show that ectopic expression of Hoxa2 in Hox-negative CNCCs results in distinct phenotypes in different CNCC subpopulations. Namely, while ectopic Hoxa2 expression is sufficient for the morphological and molecular transformation of the first PA (PA1) CNCC derivatives into the second PA (PA2)-like structures, this same genetic alteration does not provoke the transformation of derivatives of other CNCC subpopulations, but severely impairs their development. Ectopic Hoxa2 expression results in the transformation of the proximal Meckel's cartilage and of the malleus, two ventral PA1 CNCCs derivatives, into a supernumerary styloid process (SP), a PA2-derived mammalian-specific skeletal structure. These results, together with experiments to inactivate and ectopically activate the Edn1-Dlx5/6 pathway, indicate a dorsoventral PA2 (hyomandibular/ceratohyal) boundary passing through the middle of the SP. The present findings suggest context-dependent function of Hoxa2 in CNCC regional specification and morphogenesis, and provide novel insights into the evolution of taxa-specific patterning of PA-derived structures.


Subject(s)
Branchial Region/embryology , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/metabolism , Morphogenesis/physiology , Neural Crest/metabolism , Alcian Blue , Animals , Anthraquinones , Branchial Region/metabolism , DNA Primers/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental/genetics , In Situ Hybridization , Mice , Mice, Mutant Strains , Morphogenesis/genetics , Neural Crest/embryology , Real-Time Polymerase Chain Reaction
15.
EMBO J ; 31(23): 4404-14, 2012 Nov 28.
Article in English | MEDLINE | ID: mdl-23103767

ABSTRACT

Tumour necrosis factor alpha (TNFα) is a potent cytokine that signals through nuclear factor kappa B (NFκB) to activate a subset of human genes. It is usually assumed that this involves RNA polymerases transcribing responsive genes wherever they might be in the nucleus. Using primary human endothelial cells, variants of chromosome conformation capture (including 4C and chromatin interaction analysis with paired-end tag sequencing), and fluorescence in situ hybridization to detect single nascent transcripts, we show that TNFα induces responsive genes to congregate in discrete 'NFκB factories'. Some factories further specialize in transcribing responsive genes encoding micro-RNAs that target downregulated mRNAs. We expect all signalling pathways to contain this extra leg, where responding genes are transcribed in analogous specialized factories.


Subject(s)
Gene Expression Regulation , MicroRNAs/metabolism , Tumor Necrosis Factor-alpha/metabolism , Chromosomes/ultrastructure , Cytokines/biosynthesis , Cytoplasm/metabolism , DNA-Directed RNA Polymerases/metabolism , Endothelial Cells/cytology , Humans , In Situ Hybridization , In Situ Hybridization, Fluorescence , N-Acetylglucosaminyltransferases/metabolism , NF-kappa B/metabolism , Protein Conformation , Repressor Proteins/metabolism , Signal Transduction , Smad Proteins/metabolism , Time Factors , Transcription, Genetic , Transforming Growth Factor beta/metabolism
16.
Int J Mol Sci ; 17(2): 212, 2016 Feb 05.
Article in English | MEDLINE | ID: mdl-26861296

ABSTRACT

Cdkn1A-interacting zinc finger protein 1 (CIZ1) was first identified in a yeast-2-hybrid system searching for interacting proteins of CDK2 inhibitor p21(Cip1/Waf1). Ciz1 also binds to CDK2, cyclin A, cyclin E, CDC6, PCNA, TCF4 and estrogen receptor-α. Recent studies reveal numerous biological functions of CIZ1 in DNA replication, cell proliferation, and differentiation. In addition, splicing variants of CIZ1 mRNA is associated with a variety of cancers and Alzheimer's disease, and mutations of the CIZ1 gene lead to cervical dystonia. CIZ1 expression is increased in cancers and rheumatoid arthritis. In this review, we will summarize the biological functions and molecular mechanisms of CIZ1 in these physiological and pathological processes.


Subject(s)
DNA Replication , Disease Susceptibility , Nuclear Proteins/metabolism , Alternative Splicing , Animals , Cell Cycle/genetics , Cell Differentiation , Cell Proliferation , Gene Expression Regulation , Humans , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Protein Conformation
17.
EMBO J ; 30(13): 2582-95, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21666600

ABSTRACT

GATA2 is well recognized as a key transcription factor and regulator of cell-type specificity and differentiation. Here, we carried out comparative chromatin immunoprecipitation with comprehensive sequencing (ChIP-seq) to determine genome-wide occupancy of GATA2 in endothelial cells and erythroids, and compared the occupancy to the respective gene expression profile in each cell type. Although GATA2 was commonly expressed in both cell types, different GATA2 bindings and distinct cell-specific gene expressions were observed. By using the ChIP-seq with epigenetic histone modifications and chromatin conformation capture assays; we elucidated the mechanistic regulation of endothelial-specific GATA2-mediated endomucin gene expression, that was regulated by the endothelial-specific chromatin loop with a GATA2-associated distal enhancer and core promoter. Knockdown of endomucin markedly attenuated endothelial cell growth, migration and tube formation. Moreover, abrogation of GATA2 in endothelium demonstrated not only a reduction of endothelial-specific markers, but also induction of mesenchymal transition promoting gene expression. Our findings provide new insights into the correlation of endothelial-expressed GATA2 binding, epigenetic modification, and the determination of endothelial cell specificity.


Subject(s)
Endothelium, Vascular/metabolism , Epigenesis, Genetic/physiology , GATA2 Transcription Factor/metabolism , Sialoglycoproteins/genetics , Animals , Base Sequence , COS Cells , Cells, Cultured , Chlorocebus aethiops , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/drug effects , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/physiology , Gene Expression Profiling , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Knockdown Techniques , Humans , K562 Cells , Microarray Analysis , Models, Biological , Organ Specificity/drug effects , Organ Specificity/genetics , Protein Binding/genetics , Protein Binding/physiology , RNA, Small Interfering/pharmacology , Sialoglycoproteins/metabolism
18.
Proc Natl Acad Sci U S A ; 108(21): 8779-84, 2011 May 24.
Article in English | MEDLINE | ID: mdl-21555558

ABSTRACT

Diabetes and the development of its complications have been associated with mitochondrial DNA (mtDNA) dysfunction, but causal relationships remain undetermined. With the objective of testing whether increased mtDNA mutations exacerbate the diabetic phenotype, we have compared mice heterozygous for the Akita diabetogenic mutation (Akita) with mice homozygous for the D257A mutation in mitochondrial DNA polymerase gamma (Polg) or with mice having both mutations (Polg-Akita). The Polg-D257A protein is defective in proofreading and increases mtDNA mutations. At 3 mo of age, the Polg-Akita and Akita male mice were equally hyperglycemic. Unexpectedly, as the Polg-Akita males aged to 9 mo, their diabetic symptoms decreased. Thus, their hyperglycemia, hyperphagia and urine output declined significantly. The decrease in their food intake was accompanied by increased plasma leptin and decreased plasma ghrelin, while hypothalamic expression of the orexic gene, neuropeptide Y, was lower and expression of the anorexic gene, proopiomelanocortin, was higher. Testis function progressively worsened with age in the double mutants, and plasma testosterone levels in 9-mo-old Polg-Akita males were significantly reduced compared with Akita males. The hyperglycemia and hyperphagia returned in aged Polg-Akita males after testosterone administration. Hyperglycemia-associated distal tubular damage in the kidney also returned, and Polg-D257A-associated proximal tubular damage was enhanced. The mild diabetes of female Akita mice was not affected by the Polg-D257A mutation. We conclude that reduced diabetic symptoms of aging Polg-Akita males results from appetite suppression triggered by decreased testosterone associated with damage to the Leydig cells of the testis.


Subject(s)
Appetite/genetics , DNA, Mitochondrial/genetics , DNA-Directed DNA Polymerase/genetics , Diabetes Mellitus/genetics , Mice, Inbred Strains/genetics , Mutation , Aging , Animals , DNA Polymerase gamma , Diabetes Mellitus/pathology , Diabetes Mellitus/physiopathology , Female , Hyperglycemia , Leydig Cells/pathology , Male , Mice , Phenotype , RNA Editing/genetics , Testis/pathology , Testosterone/blood
19.
iScience ; 27(4): 109398, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38544573

ABSTRACT

Mitochondria play a vital role in non-shivering thermogenesis in both brown and subcutaneous white adipose tissues (BAT and scWAT, respectively). However, specific regulatory mechanisms driving mitochondrial function in these tissues have been unclear. Here we demonstrate that prolonged activation of ß-adrenergic signaling induces epigenetic modifications in scWAT, specifically targeting the enhancers for the mitochondria master regulator genes Pgc1a/b. This is mediated at least partially through JMJD1A, a histone demethylase that in response to ß-adrenergic signals, facilitates H3K9 demethylation of the Pgc1a/b enhancers, promoting mitochondrial biogenesis and the formation of beige adipocytes. Disruption of demethylation activity of JMJD1A in mice impairs activation of Pgc1a/b driven mitochondrial biogenesis and limits scWAT beiging, contributing to reduced energy expenditure, obesity, insulin resistance, and metabolic disorders. Notably, JMJD1A demethylase activity is not required for Pgc1a/b dependent thermogenic capacity of BAT especially during acute cold stress, emphasizing the importance of scWAT thermogenesis in overall energy metabolism.

20.
PLoS Biol ; 8(7): e1000419, 2010 Jul 13.
Article in English | MEDLINE | ID: mdl-20644712

ABSTRACT

It is widely assumed that active RNA polymerases track along their templates to produce a transcript. We test this using chromosome conformation capture and human genes switched on rapidly and synchronously by tumour necrosis factor alpha (TNFalpha); one is 221 kbp SAMD4A, which a polymerase takes more than 1 h to transcribe. Ten minutes after stimulation, the SAMD4A promoter comes together with other TNFalpha-responsive promoters. Subsequently, these contacts are lost as new downstream ones appear; contacts are invariably between sequences being transcribed. Super-resolution microscopy confirms that nascent transcripts (detected by RNA fluorescence in situ hybridization) co-localize at relevant times. Results are consistent with an alternative view of transcription: polymerases fixed in factories reel in their respective templates, so different parts of the templates transiently lie together.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , Base Pairing/genetics , Cells, Cultured , Chromosomes, Human, Pair 14/genetics , Enzyme Activation/drug effects , Humans , In Situ Hybridization, Fluorescence , Introns/genetics , Protein Binding/drug effects , RNA Transport/drug effects , Repressor Proteins/metabolism , Time Factors , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL