Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 91
1.
EMBO Mol Med ; 15(12): e18459, 2023 Dec 07.
Article En | MEDLINE | ID: mdl-37937685

Triple-negative breast cancer (TNBC) often develops resistance to single-agent treatment, which can be circumvented using targeted combinatorial approaches. Here, we demonstrate that the simultaneous inhibition of LOXL2 and BRD4 synergistically limits TNBC proliferation in vitro and in vivo. Mechanistically, LOXL2 interacts in the nucleus with the short isoform of BRD4 (BRD4S), MED1, and the cell cycle transcriptional regulator B-MyB. These interactions sustain the formation of BRD4 and MED1 nuclear transcriptional foci and control cell cycle progression at the gene expression level. The pharmacological co-inhibition of LOXL2 and BRD4 reduces BRD4 nuclear foci, BRD4-MED1 colocalization, and the transcription of cell cycle genes, thus suppressing TNBC cell proliferation. Targeting the interaction between BRD4S and LOXL2 could be a starting point for the development of new anticancer strategies for the treatment of TNBC.


Transcription Factors , Triple Negative Breast Neoplasms , Humans , Amino Acid Oxidoreductases/genetics , Amino Acid Oxidoreductases/metabolism , Bromodomain Containing Proteins , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Mediator Complex Subunit 1/genetics , Mediator Complex Subunit 1/metabolism , Nuclear Proteins/genetics , Transcription Factors/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Animals
2.
Mol Cell Endocrinol ; 559: 111781, 2023 01 01.
Article En | MEDLINE | ID: mdl-36191835

Thyroid hormone receptor (TR) controls the expression of thyroid hormone (T3)-responsive genes, while undergoing rapid nucleocytoplasmic shuttling. In Resistance to Thyroid Hormone syndrome (RTH), mutant TR fails to activate T3-dependent transcription. Previously, we showed that Mediator subunit 1 (MED1) plays a role in TR nuclear retention. Here, we investigated MED1's effect on RTH mutants using nucleocytoplasmic scoring and fluorescence recovery after photobleaching in transfected cells. MED1 overexpression and knockout did not change the nucleocytoplasmic distribution or intranuclear mobility of C392X and P398R TRα1 at physiological T3 levels. At elevated T3 levels, however, overexpression increased P398R's nuclear retention and MED1 knockout decreased P398R's and A263V's intranuclear mobility, while not impacting C392X. Although A263V TRα1-transfected cells had a high percentage of aggregates, MED1 rescued A263V's impaired intranuclear mobility, suggesting that MED1 ameliorates nonfunctional aggregates. Results correlate with clinical severity, suggesting that altered interaction between MED1 and TRα1 mutants contributes to RTH pathology.


Receptors, Thyroid Hormone , Thyroid Hormone Resistance Syndrome , Humans , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/metabolism , Mutation/genetics , Thyroid Hormone Resistance Syndrome/genetics , Cell Nucleus/metabolism , Thyroid Hormones/metabolism , Triiodothyronine/metabolism , Mediator Complex Subunit 1/genetics , Mediator Complex Subunit 1/metabolism
3.
Sci Rep ; 12(1): 17140, 2022 10 13.
Article En | MEDLINE | ID: mdl-36229463

Homologous recombination (HR) is a major repair pathway of DNA double-strand breaks and is closely related to carcinogenesis. HR deficiency has been established as a therapeutic target. The aim of this study was to elucidate the functions of a novel HR factor, Mediator complex subunit 1 (MED1), and its association with BRCA1. Formation of the MED1/BRCA1 complex was examined by immunoprecipitation and GST-pull down assays. The transcription cofactor role of BRCA1 was evaluated using luciferase assays. The roles of MED1 on DNA damage response and HR were analyzed by immunofluorescence and HR assays. R-loop accumulation was analyzed using immunofluorescence. R-loop-induced DNA damage was analyzed by comet assays. Immunoprecipitation and GST-pull down assays demonstrated that MED1 is a novel binding partner of BRCA1 and binds to the BRCT domain. Luciferase assays showed that MED1 potentiated the transcription ability of BRCT by two-fold. In MED1-depleted cells, recruitment of HR genes, such as RPA and γH2AX, to DNA damage sites was severely impaired. HR assays showed that MED1 knockdown significantly decreased HR activity. R-loop nuclear accumulation and R-loop-induced comet tails were observed in MED1-depleted cells. We conclude that the transcription factor MED1 contributes to the regulation of the HR pathway and R-loop processing.


Mediator Complex Subunit 1 , R-Loop Structures , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , DNA , DNA Repair , Homologous Recombination , Mediator Complex Subunit 1/genetics , Transcription Factors/metabolism
4.
Circ Res ; 131(10): 828-841, 2022 10 28.
Article En | MEDLINE | ID: mdl-36252121

BACKGROUND: Dysregulated BMP (bone morphogenetic protein) or TGF-ß (transforming growth factor beta) signaling pathways are imperative in idiopathic and familial pulmonary arterial hypertension (PAH) as well as experimental pulmonary hypertension (PH) in rodent models. MED1 (mediator complex subunit 1) is a key transcriptional co-activator and KLF4 (Krüppel-like factor 4) is a master transcription factor in endothelium. However, MED1 and KLF4 epigenetic and transcriptional regulations of the BMP/TGF-ß axes in pulmonary endothelium and their dysregulations leading to PAH remain elusive. We investigate the MED1/KLF4 co-regulation of the BMP/TGF-ß axes in endothelium by studying the epigenetic regulation of BMPR2 (BMP receptor type II), ETS-related gene (ERG), and TGFBR2 (TGF-ß receptor 2) and their involvement in the PH. METHODS: High-throughput screening involving data from RNA-seq, MED1 ChIP-seq, H3K27ac ChIP-seq, ATAC-seq, and high-throughput chromosome conformation capture together with in silico computations were used to explore the epigenetic and transcriptional regulation of BMPR2, ERG, and TGFBR2 by MED1 and KLF4. In vitro experiments with cultured pulmonary arterial endothelial cells (ECs) and bulk assays were used to validate results from these in silico analyses. Lung tissue from patients with idiopathic PAH, animals with experimental PH, and mice with endothelial ablation of MED1 (EC-MED1-/-) were used to study the PH-protective effect of MED1. RESULTS: Levels of MED1 were decreased in lung tissue or pulmonary arterial endothelial cells from idiopathic PAH patients and rodent PH models. Mechanistically, MED1 acted synergistically with KLF4 to transactivate BMPR2, ERG, and TGFBR2 via chromatin remodeling and enhancer-promoter interactions. EC-MED1-/- mice showed PH susceptibility. In contrast, MED1 overexpression mitigated the PH phenotype in rodents. CONCLUSIONS: A homeostatic regulation of BMPR2, ERG, and TGFBR2 in ECs by MED1 synergistic with KLF4 is essential for the normal function of the pulmonary endothelium. Dysregulation of MED1 and the resulting impairment of the BMP/TGF-ß signaling is implicated in the disease progression of PAH in humans and PH in rodent models.


Hypertension, Pulmonary , Pulmonary Arterial Hypertension , Humans , Mice , Animals , Hypertension, Pulmonary/metabolism , Transforming Growth Factor beta/metabolism , Receptor, Transforming Growth Factor-beta Type II/genetics , Endothelial Cells/metabolism , Epigenesis, Genetic , Bone Morphogenetic Protein Receptors, Type II/genetics , Bone Morphogenetic Protein Receptors, Type II/metabolism , Pulmonary Artery/metabolism , Bone Morphogenetic Proteins/genetics , Pulmonary Arterial Hypertension/genetics , Endothelium, Vascular/metabolism , Transcription Factors/metabolism , Mediator Complex Subunit 1/genetics , Mediator Complex Subunit 1/metabolism
5.
Head Neck ; 44(11): 2417-2427, 2022 11.
Article En | MEDLINE | ID: mdl-35916453

BACKGROUND: The potential role of circFNDC3B in regulating oral tongue squamous cell carcinoma development (OTSCC) remains unknown. METHODS: The level of circFNDC3B in OTSCC tissues or cell lines was measured and its function in vitro and in vivo was analyzed. Interactions among circFNDC3B, miR-1322, and MED1 were verified by luciferase reporter and RNA pull-down assays. RESULTS: The level of circFNDC3B in tissues or cell lines of OTSCC was higher than that in control groups. siRNA-mediated circFNDC3B inhibition resulted in weakened proliferation, migration, and invasion, which was reversed by miR-1322. Overexpression of MED1 in OTSCC cells partially reversed the tumor suppression functions of si-circFNDC3B or miR-1322 mimics in vitro. circFNDC3B overexpression dramatically promoted tumor growth in vivo. circFNDC3B directly bound with miR-1322 and consequently promoted the MED1 expression in OTSCC cells. CONCLUSIONS: The circFNDC3B/miR-1322/MED1 axis participates in OTSCC progression, which may provide novel therapeutic targets for OTSCC.


MicroRNAs , RNA, Circular , Squamous Cell Carcinoma of Head and Neck , Tongue Neoplasms , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Fibronectins/genetics , Gene Expression Regulation, Neoplastic , Humans , Mediator Complex Subunit 1/genetics , Mediator Complex Subunit 1/metabolism , MicroRNAs/genetics , RNA, Circular/genetics , RNA, Small Interfering , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/pathology , Tongue Neoplasms/genetics , Tongue Neoplasms/pathology
6.
Am J Pathol ; 192(7): 1016-1027, 2022 07.
Article En | MEDLINE | ID: mdl-35461855

Mediator 1 (MED1), a key subunit of the mediator complex, interacts with various nuclear receptors and functions in lipid metabolism and energy homeostasis. Dilated cardiomyopathy-related ventricular dilatation and heart failure have been reported in mice with cardiomyocyte-specific Med1 deficiency. However, the contribution of macrophage-specific MED1 in cardiac remodeling remains unclear. In this study, macrophage-specific Med1 knockout (Med1ΔMac) mice were generated and exposed to isoproterenol (ISO) to induce cardiac fibrosis; these mice showed aggravated cardiac fibrosis compared with Med1fl/fl mice. The levels of expression of marker genes for myofibroblast transdifferentiation [α-smooth muscle actin (SMA)] and of profibrotic genes, including Col1a1, Col3a1, Postn, Mmp2, Timp1, and Fn1, were significantly increased in the cardiac tissues of Med1ΔMac mice with ISO-induced myocardial fibrosis. In particular, the transforming growth factor (TGF)-ß-Smad2/3 signaling pathway was activated. In bone marrow-derived and peritoneal macrophages, Med1 deficiency was also associated with elevated levels of expression of proinflammatory genes, including Il6, Tnfa, and Il1b. These findings indicate that macrophage-specific MED1 deficiency may aggravate ISO-induced cardiac fibrosis via the regulation of the TGF-ß-SMAD2/3 pathway, and the underlying mechanism may involve MED1 deficiency triggering the activation of inflammatory cytokines in macrophages, which in turn may stimulate phenotypic switch of cardiac fibroblasts and accelerate cardiac fibrosis. Thus, MED1 is a potential therapeutic target for cardiac fibrosis.


Isoproterenol , Macrophages , Mediator Complex Subunit 1 , Myocytes, Cardiac , Animals , Fibrosis , Isoproterenol/toxicity , Macrophages/metabolism , Mediator Complex Subunit 1/deficiency , Mediator Complex Subunit 1/genetics , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/pathology , Myofibroblasts/metabolism
7.
Nucleic Acids Res ; 50(8): 4450-4463, 2022 05 06.
Article En | MEDLINE | ID: mdl-35394046

Mediator activates RNA polymerase II (Pol II) function during transcription, but it remains unclear whether Mediator is able to travel with Pol II and regulate Pol II transcription beyond the initiation and early elongation steps. By using in vitro and in vivo transcription recycling assays, we find that human Mediator 1 (MED1), when phosphorylated at the mammal-specific threonine 1032 by cyclin-dependent kinase 9 (CDK9), dynamically moves along with Pol II throughout the transcribed genes to drive Pol II recycling after the initial round of transcription. Mechanistically, MED31 mediates the recycling of phosphorylated MED1 and Pol II, enhancing mRNA output during the transcription recycling process. Importantly, MED1 phosphorylation increases during prostate cancer progression to the lethal phase, and pharmacological inhibition of CDK9 decreases prostate tumor growth by decreasing MED1 phosphorylation and Pol II recycling. Our results reveal a novel role of MED1 in Pol II transcription and identify phosphorylated MED1 as a targetable driver of dysregulated Pol II recycling in cancer.


Neoplasms , RNA Polymerase II , Animals , Humans , Male , Mammals/genetics , Mediator Complex/metabolism , Mediator Complex Subunit 1/genetics , Neoplasms/genetics , Phosphorylation , RNA Polymerase II/metabolism , Transcription, Genetic
8.
J Invest Dermatol ; 142(8): 2228-2237.e4, 2022 08.
Article En | MEDLINE | ID: mdl-35131256

Metastasis is the main reason for the high mortality of patients and indeed a difficult task in the treatment of cutaneous melanoma. Therefore, it is of great clinical value to explore the molecular mechanism of cutaneous metastatic melanoma and develop novel therapies. MED1, acting as a factor required for activator-dependent transcription, is reported to be involved in carcinogenesis and progression. In this study, we found that MED1 was highly expressed in patients with cutaneous melanoma. MED1 downregulation could induce cellular epithelial-to-mesenchymal transition and promote migration, invasion, and metastasis of cutaneous melanoma in vivo and in vitro. Further analysis showed that in Med1 knockdown cells, the TGFß/SMAD2 signaling pathway mediated an increase in epithelial-to-mesenchymal transition phenotype and migration. The opposite results were observed after treatment with TGFß inhibitors. To further explore the mechanism, we found that MED1 interacted with SMAD2, and MED1 downregulation could protect SMAD2 from degradation by inhibiting SMAD2 ubiquitination. Together, these results suggest that MED1 inhibited TGFß signaling pathway to reduce cell epithelial-to-mesenchymal transition phenotype and migration through SMAD2 ubiquitination in the metastasis of cutaneous melanoma. Our findings elucidated the role of MED1 in the metastasis of cutaneous melanoma and provided a target for the therapeutic strategies of cutaneous melanoma.


Mediator Complex Subunit 1 , Melanoma , Skin Neoplasms , Smad2 Protein , Cell Line, Tumor , Cell Movement/genetics , Down-Regulation , Epithelial-Mesenchymal Transition , Humans , Mediator Complex Subunit 1/genetics , Mediator Complex Subunit 1/metabolism , Melanoma/pathology , Signal Transduction/genetics , Skin Neoplasms/pathology , Smad2 Protein/metabolism , Transforming Growth Factor beta/metabolism , Ubiquitination , Melanoma, Cutaneous Malignant
9.
Nucleic Acids Res ; 50(1): 207-226, 2022 01 11.
Article En | MEDLINE | ID: mdl-34931241

CTCF is crucial to the organization of mammalian genomes into loop structures. According to recent studies, the transcription apparatus is compartmentalized and concentrated at super-enhancers to form phase-separated condensates and drive the expression of cell-identity genes. However, it remains unclear whether and how transcriptional condensates are coupled to higher-order chromatin organization. Here, we show that CTCF is essential for RNA polymerase II (Pol II)-mediated chromatin interactions, which occur as hyperconnected spatial clusters at super-enhancers. We also demonstrate that CTCF clustering, unlike Pol II clustering, is independent of liquid-liquid phase-separation and resistant to perturbation of transcription. Interestingly, clusters of Pol II, BRD4, and MED1 were found to dissolve upon CTCF depletion, but were reinstated upon restoration of CTCF, suggesting a potent instructive function for CTCF in the formation of transcriptional condensates. Overall, we provide evidence suggesting that CTCF-mediated chromatin looping acts as an architectural prerequisite for the assembly of phase-separated transcriptional condensates.


CCCTC-Binding Factor/metabolism , Chromatin Assembly and Disassembly , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , Epigenesis, Genetic , HCT116 Cells , Humans , Mediator Complex Subunit 1/genetics , Mediator Complex Subunit 1/metabolism , RNA Polymerase II/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Bioengineered ; 12(1): 7600-7615, 2021 12.
Article En | MEDLINE | ID: mdl-34612778

Liver failure has attracted attention in clinical work due to its high mortality, and the development of liver transplantation is restricted by various factors. Therefore, it is very important to carry out research on the mechanism of liver cell regeneration. This article has studied in depth the preparation of MED1 gene nanocarriers, collected human plasmids and cells through experimental materials and experimental instruments, and conducted comparative research on conventional culture. This question conducts a regeneration experiment on liver cells in chronic-onset acute liver failure, divides patients into an experimental group and a control group, and understands the recovery of liver function according to the screening of their plasma samples and separation of plasma. This article selects the commonly used clinical biological markers, such as Na+, AFP, Alb, CHE (serum cholinesterase) and other indicators to reflect the regeneration ability of liver function. The incidence of surgical complications in the control group, such as ascites, infection, bleeding, HE, hepatorenal syndrome, and hyponatremia were 71.3%, 87.4%, 16.1%, 41.4%, 19.5%, and 33.3%, respectively. Significantly higher than the experimental group, the difference was statistically significant (P < 0.05); while gender, age, PLT level and whether to use hormones, artificial liver or not there was no significant difference between the two groups (P > 0.05).


Gene Transfer Techniques , Liver Failure, Acute , Liver Regeneration/genetics , Mediator Complex Subunit 1 , Nanostructures , Adult , Cells, Cultured , Female , Hepatocytes/cytology , Humans , Liver/cytology , Liver/metabolism , Male , Mediator Complex Subunit 1/genetics , Mediator Complex Subunit 1/metabolism , Middle Aged , Plasmids/genetics
12.
Nat Commun ; 12(1): 3974, 2021 06 25.
Article En | MEDLINE | ID: mdl-34172737

Cancer stem cells (CSCs) play a critical role in invasive growth and metastasis of human head and neck squamous cell carcinoma (HNSCC). Although significant progress has been made in understanding the self-renewal and pro-tumorigenic potentials of CSCs, a key challenge remains on how to eliminate CSCs and halt metastasis effectively. Here we show that super-enhancers (SEs) play a critical role in the transcription of cancer stemness genes as well as pro-metastatic genes, thereby controlling their tumorigenic potential and metastasis. Mechanistically, we find that bromodomain-containing protein 4 (BRD4) recruits Mediators and NF-κB p65 to form SEs at cancer stemness genes such as TP63, MET and FOSL1, in addition to oncogenic transcripts. In vivo lineage tracing reveals that disrupting SEs by BET inhibitors potently inhibited CSC self-renewal and eliminated CSCs in addition to elimination of proliferating non-stem tumor cells in a mouse model of HNSCC. Moreover, disrupting SEs also inhibits the invasive growth and lymph node metastasis of human CSCs isolated from human HNSCC. Taken together, our results suggest that targeting SEs may serve as an effective therapy for HNSCC by eliminating CSCs.


Enhancer Elements, Genetic , Head and Neck Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Squamous Cell Carcinoma of Head and Neck/pathology , Animals , Antineoplastic Agents/pharmacology , Azepines/pharmacology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Humans , Lymphatic Metastasis/drug therapy , Lymphatic Metastasis/prevention & control , Mediator Complex Subunit 1/genetics , Mediator Complex Subunit 1/metabolism , Mice, Inbred C57BL , Mice, SCID , NF-kappa B/genetics , Neoplastic Stem Cells/pathology , Polycomb Repressive Complex 1/antagonists & inhibitors , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Triazoles/pharmacology , Xenograft Model Antitumor Assays
13.
IUBMB Life ; 73(9): 1142-1152, 2021 09.
Article En | MEDLINE | ID: mdl-34117711

Macrophage polarization is of great importance in the formation of atherosclerotic plaque. Homeobox A5 (HOXA5), one of the homeobox transcription factors, has been revealed to be closely associated with macrophage phenotype switching. This study aims to investigate the role of HOXA5 in carotid atherosclerosis (CAS). Herein, the role of HOXA5 was explored in polarized RAW264.7 macrophages in vitro and ApoE-/- mice in vivo. Interestingly, compared with that in M0 macrophages, both the mRNA and protein expression levels of HOXA5 were decreased in lipopolysaccharide (LPS)/interferon (IFN)-γ-induced M1 macrophages, while increased in IL-4-induced M2 macrophages. In addition, in the presence of IL-4, HOXA5-overexpressing RAW264.7 cells preferred to polarizing toward M2 phenotypes. Furthermore, we found that HOXA5 bound to the promoter region and activated the expression of mediator subunit 1 (MED1), a gene known to regulate macrophage differentiation. Knocking MED1 down inhibited HOXA5-enhanced M2 macrophage polarization. In vivo, the CAS model was induced in ApoE-/- mouse fed with a Western-type diet and placed a perivascular carotid collar. Decreased mRNA and protein expressions of HOXA5 were observed in carotid arteries of CAS mice. Forced overexpression of HOXA5 reduced intimal hyperplasia and lipid accumulation in carotid vessels, and it also promoted the polarization of macrophages to M2 subtypes. The expression of MED1 was decreased in atherosclerotic carotid vessels, while HOXA5 overexpression restored its change. Collectively, HOXA5 in carotid arteries is involved in the macrophage M1/M2 switching in atherosclerotic plaque, which may be associated with its transcriptional regulation of MED1.


Carotid Artery Diseases/physiopathology , Homeodomain Proteins/metabolism , Macrophages/physiology , Mediator Complex Subunit 1/metabolism , Transcription Factors/metabolism , Animals , Carotid Artery Diseases/genetics , Cell Polarity , Down-Regulation , Gene Expression Regulation , Homeodomain Proteins/genetics , Macrophages/pathology , Male , Mediator Complex Subunit 1/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/pathology , RAW 264.7 Cells , Transcription Factors/genetics
14.
Genes Dev ; 35(9-10): 713-728, 2021 05 01.
Article En | MEDLINE | ID: mdl-33888555

MED1 often serves as a surrogate of the general transcription coactivator complex Mediator for identifying active enhancers. MED1 is required for phenotypic conversion of fibroblasts to adipocytes in vitro, but its role in adipose development and expansion in vivo has not been reported. Here, we show that MED1 is not generally required for transcription during adipogenesis in culture and that MED1 is dispensable for adipose development in mice. Instead, MED1 is required for postnatal adipose expansion and the induction of fatty acid and triglyceride synthesis genes after pups switch diet from high-fat maternal milk to carbohydrate-based chow. During adipogenesis, MED1 is dispensable for induction of lineage-determining transcription factors (TFs) PPARγ and C/EBPα but is required for lipid accumulation in the late phase of differentiation. Mechanistically, MED1 controls the induction of lipogenesis genes by facilitating lipogenic TF ChREBP- and SREBP1a-dependent recruitment of Mediator to active enhancers. Together, our findings identify a cell- and gene-specific regulatory role of MED1 as a lipogenesis coactivator required for postnatal adipose expansion.


Adipose Tissue/growth & development , Gene Expression Regulation, Developmental/genetics , Lipogenesis/genetics , Mediator Complex Subunit 1/genetics , Mediator Complex Subunit 1/metabolism , Adipose Tissue/metabolism , Adipose Tissue, Brown/embryology , Animals , Cells, Cultured , Diet , Mice , Protein Binding/genetics
15.
Genes Dev ; 35(9-10): 729-748, 2021 05 01.
Article En | MEDLINE | ID: mdl-33888560

The MED1 subunit has been shown to mediate ligand-dependent binding of the Mediator coactivator complex to multiple nuclear receptors, including the adipogenic PPARγ, and to play an essential role in ectopic PPARγ-induced adipogenesis of mouse embryonic fibroblasts. However, the precise roles of MED1, and its various domains, at various stages of adipogenesis and in adipose tissue have been unclear. Here, after establishing requirements for MED1, including specific domains, for differentiation of 3T3L1 cells and both primary white and brown preadipocytes, we used multiple genetic approaches to assess requirements for MED1 in adipocyte formation, maintenance, and function in mice. We show that MED1 is indeed essential for the differentiation and/or function of both brown and white adipocytes, as its absence in these cells leads to, respectively, defective brown fat function and lipodystrophy. This work establishes MED1 as an essential transcriptional coactivator that ensures homeostatic functions of adipocytes.


Adipocytes/cytology , Cell Differentiation/genetics , Gene Expression Regulation, Developmental/genetics , Mediator Complex Subunit 1/genetics , Mediator Complex Subunit 1/metabolism , 3T3-L1 Cells , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Cells, Cultured , Embryonic Stem Cells/cytology , Mediator Complex/genetics , Mice , Protein Binding/genetics , Protein Domains
16.
Cell Rep ; 34(10): 108822, 2021 03 09.
Article En | MEDLINE | ID: mdl-33691110

MED1 (mediator subunit 1) co-amplifies with HER2, but its role in HER2-driven mammary tumorigenesis is still unknown. Here, we generate MED1 mammary-specific overexpression mice and cross them with mouse mammary tumor virus (MMTV)-HER2 mice. We observe significantly promoted onset, growth, metastasis, and multiplicity of HER2 tumors by MED1 overexpression. Further studies reveal critical roles for MED1 in epithelial-mesenchymal transition, cancer stem cell formation, and response to anti-HER2 therapy. Mechanistically, RNA sequencing (RNA-seq) transcriptome analyses and clinical sample correlation studies identify Jab1, a component of the COP9 signalosome complex, as the key direct target gene of MED1 contributing to these processes. Further studies reveal that Jab1 can also reciprocally regulate the stability and transcriptional activity of MED1. Together, our findings support a functional cooperation between these co-amplified genes in HER2+ mammary tumorigenesis and their potential usage as therapeutic targets for the treatment of HER2+ breast cancers.


Mammary Neoplasms, Experimental/pathology , Mediator Complex Subunit 1/metabolism , Receptor, ErbB-2/metabolism , Animals , Antineoplastic Agents/therapeutic use , COP9 Signalosome Complex/antagonists & inhibitors , COP9 Signalosome Complex/genetics , COP9 Signalosome Complex/metabolism , Cell Movement , Epithelial-Mesenchymal Transition , Female , Genetic Vectors/genetics , Genetic Vectors/metabolism , Lapatinib/therapeutic use , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/genetics , Mammary Tumor Virus, Mouse/genetics , Mediator Complex Subunit 1/genetics , Mice , Mice, Nude , Neoplasm Metastasis , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Phenotype , RNA Interference , RNA, Small Interfering/metabolism , Transcriptional Activation
17.
FASEB J ; 35(4): e21180, 2021 04.
Article En | MEDLINE | ID: mdl-33715222

Liver ischemia-reperfusion (I/R) injury is a common clinical pathological phenomenon, which is accompanied by the occurrence in liver transplantation. However, the underlying mechanism is not yet fully understood. MicroRNAs (miRNAs) play an important role in liver I/R injury. Therefore, the study of miRNAs function will contribute a new biological marker diagnosis of liver I/R injury. This study aims to evaluate effects of miR-497-5p in liver I/R injury in mice. The related regulatory factors of miR-497-5p in liver I/R injury were predicted by bioinformatics analysis. Vascular occlusion was performed to establish the liver I/R injury animal models. Hypoxia/reoxygenation (H/R) was performed to establish the in vitro models. Hematoxylin-eosin (HE) staining was conducted to assess liver injury. The inflammatory factors were evaluated by enzyme-linked immunosorbent assay (ELISA). Flow cytometry was adopted to assess the cell apoptosis. The expression of miR-497b-5p was increased in liver I/R injury. Knockdown of miR-497b-5p inhibited the production of inflammatory factors and cell apoptosis. Overexpression of mediator complex subunit 1 (MED1) and tissue inhibitor of metalloproteinase 2 (TIMP2) inhibited cell apoptosis to alleviate liver I/R injury. miR-497b-5p could activate the nuclear factor kappa-B (NF-κB) pathway by inhibiting the MED1/TIMP-2 axis to promote liver I/R injury. This study may provide a new strategy for the treatment of liver I/R injury.


Curcumin/pharmacology , Liver Diseases/etiology , Mediator Complex Subunit 1/metabolism , MicroRNAs/antagonists & inhibitors , Reperfusion Injury/pathology , Tissue Inhibitor of Metalloproteinase-2/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Gene Expression Regulation/drug effects , Hepatocytes , Kupffer Cells , Liver Diseases/metabolism , Male , Mediator Complex Subunit 1/genetics , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Oxygen , Tissue Inhibitor of Metalloproteinase-2/genetics
18.
J Cell Mol Med ; 25(10): 4870-4876, 2021 05.
Article En | MEDLINE | ID: mdl-33733611

Under steady-state conditions, the pool size of peripheral CD8+ T cells is maintained through turnover and survival. Beyond TCR and IL-7R signals, the underlying mechanisms are less well understood. In the present study, we found a significant reduction of CD8+ T cell proportion in spleens but not in thymi of mice with T cell-specific deletion of Mediator Subunit 1 (Med1). A competitive transfer of wild-type (WT) and Med1-deficient CD8+ T cells reproduced the phenotype in the same recipients and confirmed intrinsic role of Med1. Furthermore, we observed a comparable degree of migration and proliferation but a significant increase of cell death in Med1-deficient CD8+ T cells compared with WT counterparts. Finally, Med1-deficient CD8+ T cells exhibited a decreased expression of interleukin-7 receptor α (IL-7Rα), down-regulation of phosphorylated-STAT5 (pSTAT5) and Bim up-regulation. Collectively, our study reveals a novel role of Med1 in the maintenance of CD8+ T cells through IL-7Rα/STAT5 pathway-mediated cell survival.


CD8-Positive T-Lymphocytes , Mediator Complex Subunit 1/immunology , Receptors, Interleukin-7/immunology , Spleen/immunology , Animals , Apoptosis , Bone Marrow Cells , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Cell Survival , Cells, Cultured , Mediator Complex Subunit 1/genetics , Mice , Mice, Knockout , Mice, Transgenic , Signal Transduction , Spleen/cytology
19.
Autophagy ; 17(12): 4043-4061, 2021 12.
Article En | MEDLINE | ID: mdl-33734012

Hepatic macroautophagy/autophagy and fatty acid metabolism are transcriptionally regulated by nuclear receptors (NRs); however, it is not known whether their transcriptional co-activators are involved in autophagy. We thus examined MED1 (mediator complex subunit 1), a key component of the Mediator Complex that directly interacts with NRs, on these processes. We found that MED1 knockdown (KD) in cultured hepatic cells decreased autophagy and mitochondrial activity that was accompanied by decreased transcription of genes involved in these processes. Lipophagy and fatty acid ß-oxidation also were impaired. These effects also occurred after thyroid hormone stimulation, nutrient-replete or -deplete conditions, and in liver-specific Med1 KD (Med1 LKD) mice under fed and fasting conditions. Together, these findings showed that Med1 played a key role in hepatic autophagy, mitochondria function, and lipid metabolism under these conditions. Additionally, we identified downregulated hepatic genes in Med1 LKD mice, and subjected them to ChIP Enrichment Analysis. Our findings showed that the transcriptional activity of several NRs and transcription factors (TFs), including PPARA and FOXO1, likely were affected by Med1 LKD. Finally, Med1 expression and autophagy also were decreased in two mouse models of nonalcoholic fatty liver disease (NAFLD) suggesting that decreased Med1 may contribute to hepatosteatosis. In summary, MED1 plays an essential role in regulating hepatic autophagy and lipid oxidation during different hormonal and nutrient conditions. Thus, MED1 may serve as an integrator of multiple transcriptional pathways involved in these metabolic processes.Abbreviations: BAF: bafilomycin A1; db/db mice; Leprdb/db mice; ECAR: extracellular acidification rate; KD: knockdown; MED1: mediator complex subunit 1; NAFLD: nonalcoholic fatty liver disease; OCR: oxygen consumption rate; PPARA/PPARα: peroxisomal proliferator activated receptor alpha; TF: transcription factor; TFEB: transcription factor EB; tf-LC3: tandem fluorescence RFP-GFP-LC3; TG: triglyceride; TH: Thyroid hormone; TR: thyroid hormone receptors; V-ATPase: vacuolar-type H+-ATPase; WDF: Western diet with 15% fructose in drinking water.


Lipid Metabolism , Non-alcoholic Fatty Liver Disease , Animals , Autophagy/genetics , Lipid Metabolism/genetics , Liver/metabolism , Mediator Complex Subunit 1/genetics , Mediator Complex Subunit 1/metabolism , Mice , Non-alcoholic Fatty Liver Disease/genetics , PPAR alpha/metabolism
20.
Proc Natl Acad Sci U S A ; 118(9)2021 03 02.
Article En | MEDLINE | ID: mdl-33622787

HLA-C arose during evolution of pregnancy in the great apes 10 to 15 million years ago. It has a dual function on placental extravillous trophoblasts (EVTs) as it contributes to both tolerance and immunity at the maternal-fetal interface. The mode of its regulation is of considerable interest in connection with the biology of pregnancy and pregnancy abnormalities. First-trimester primary EVTs in which HLA-C is highly expressed, as well as JEG3, an EVT model cell line, were employed. Single-cell RNA-seq data and quantitative PCR identified high expression of the transcription factor ELF3 in those cells. Chromatin immunoprecipitation (ChIP)-PCR confirmed that both ELF3 and MED1 bound to the proximal HLA-C promoter region. However, binding of RFX5 to this region was absent or severely reduced, and the adjacent HLA-B locus remained closed. Expression of HLA-C was inhibited by ELF3 small interfering RNAs (siRNAs) and by wrenchnolol treatment. Wrenchnolol is a cell-permeable synthetic organic molecule that mimics ELF3 and is relatively specific for binding to ELF3's coactivator, MED23, as our data also showed in JEG3. Moreover, the ELF3 gene is regulated by a superenhancer that spans more than 5 Mb, identified by assay for transposase-accessible chromatin using sequencing (ATAC-seq), as well as by its sensitivity to (+)-JQ1 (inhibitor of BRD4). ELF3 bound to its own promoter, thus creating an autoregulatory feedback loop that establishes expression of ELF3 and HLA-C in trophoblasts. Wrenchnolol blocked binding of MED23 to ELF3, thus disrupting the positive-feedback loop that drives ELF3 expression, with down-regulation of HLA-C expression as a consequence.


DNA-Binding Proteins/genetics , Enhancer Elements, Genetic , Feedback, Physiological , HLA-C Antigens/genetics , Proto-Oncogene Proteins c-ets/genetics , Transcription Factors/genetics , Trophoblasts/immunology , Abortion, Legal , Adamantane/pharmacology , Azepines/pharmacology , Cell Line , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/immunology , Female , Gene Expression Regulation, Developmental/immunology , HLA-B Antigens/genetics , HLA-B Antigens/immunology , HLA-C Antigens/immunology , Humans , Immunity, Maternally-Acquired , Indoles/pharmacology , Mediator Complex/genetics , Mediator Complex/immunology , Mediator Complex Subunit 1/genetics , Mediator Complex Subunit 1/immunology , Pregnancy , Pregnancy Trimester, First , Primary Cell Culture , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-ets/antagonists & inhibitors , Proto-Oncogene Proteins c-ets/immunology , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , Regulatory Factor X Transcription Factors/genetics , Regulatory Factor X Transcription Factors/immunology , Signal Transduction , Transcription Factors/antagonists & inhibitors , Transcription Factors/immunology , Triazoles/pharmacology , Trophoblasts/cytology , Trophoblasts/drug effects
...