Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Proc Natl Acad Sci U S A ; 117(9): 4792-4801, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32075915

ABSTRACT

Angiogenesis, the formation of new blood vessels, is tightly regulated by gene transcriptional programs. Yin Ying 1 (YY1) is a ubiquitously distributed transcription factor with diverse and complex biological functions; however, little is known about the cell-type-specific role of YY1 in vascular development and angiogenesis. Here we report that endothelial cell (EC)-specific YY1 deletion in mice led to embryonic lethality as a result of abnormal angiogenesis and vascular defects. Tamoxifen-inducible EC-specific YY1 knockout (YY1iΔEC ) mice exhibited a scarcity of retinal sprouting angiogenesis with fewer endothelial tip cells. YY1iΔEC mice also displayed severe impairment of retinal vessel maturation. In an ex vivo mouse aortic ring assay and a human EC culture system, YY1 depletion impaired endothelial sprouting and migration. Mechanistically, YY1 functions as a repressor protein of Notch signaling that controls EC tip-stalk fate determination. YY1 deficiency enhanced Notch-dependent gene expression and reduced tip cell formation. Specifically, YY1 bound to the N-terminal domain of RBPJ (recombination signal binding protein for Ig Kappa J region) and competed with the Notch coactivator MAML1 (mastermind-like protein 1) for binding to RBPJ, thereby impairing the NICD (intracellular domain of the Notch protein)/MAML1/RBPJ complex formation. Our study reveals an essential role of endothelial YY1 in controlling sprouting angiogenesis through directly interacting with RBPJ and forming a YY1-RBPJ nuclear repression complex.


Subject(s)
Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Morphogenesis/physiology , Neovascularization, Pathologic/metabolism , YY1 Transcription Factor/metabolism , Animals , Carrier Proteins/metabolism , Cell Differentiation , Endothelial Cells/metabolism , Female , Gene Expression Regulation, Developmental , Humans , Male , Mice/embryology , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/physiology , Nuclear Proteins , Protein Binding , Receptors, Notch/metabolism , Retinal Vessels/metabolism , Signal Transduction , Transcription Factors , YY1 Transcription Factor/genetics
2.
Hepatology ; 72(5): 1717-1734, 2020 11.
Article in English | MEDLINE | ID: mdl-32048304

ABSTRACT

BACKGROUND AND AIMS: Liver fibrosis (LF) is a central pathological process that occurs in most types of chronic liver diseases. Advanced LF causes cirrhosis, hepatocellular carcinoma, and liver failure. However, the exact molecular mechanisms underlying the initiation and progression of LF remain largely unknown. APPROACH AND RESULTS: This study was designed to investigate the role of protein kinase D3 (PKD3; gene name Prkd3) in the regulation of liver homeostasis. We generated global Prkd3 knockout (Prkd3-/- ) mice and myeloid-cell-specific Prkd3 knockout (Prkd3∆LysM ) mice, and we found that both Prkd3-/- mice and Prkd3∆LysM mice displayed spontaneous LF. PKD3 deficiency also aggravated CCl4 -induced LF. PKD3 is highly expressed in hepatic macrophages (HMs), and PKD3 deficiency skewed macrophage polarization toward a profibrotic phenotype. Activated profibrotic macrophages produced transforming growth factor beta that, in turn, activates hepatic stellate cells to become matrix-producing myofibroblasts. Moreover, PKD3 deficiency decreased the phosphatase activity of SH2-containing protein tyrosine phosphatase-1 (a bona-fide PKD3 substrate), resulting in sustained signal transducer and activator of transcription 6 activation in macrophages. In addition, we observed that PKD3 expression in HMs was down-regulated in cirrhotic human liver tissues. CONCLUSIONS: PKD3 deletion in mice drives LF through the profibrotic macrophage activation.


Subject(s)
Liver Cirrhosis, Experimental/pathology , Liver Cirrhosis/pathology , Protein Kinase C/deficiency , Animals , Carbon Tetrachloride/toxicity , Cells, Cultured , Disease Progression , Hepatic Stellate Cells/metabolism , Humans , Liver/cytology , Liver/pathology , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/diagnosis , Liver Cirrhosis, Experimental/genetics , Macrophages/metabolism , Mice , Mice, Knockout , Myofibroblasts/metabolism , Primary Cell Culture , Protein Kinase C/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Severity of Illness Index , Tissue Array Analysis , Transforming Growth Factor beta/metabolism
3.
Biochem Biophys Res Commun ; 521(2): 279-284, 2020 01 08.
Article in English | MEDLINE | ID: mdl-31668369

ABSTRACT

Serum response factor (SRF), a key transcription factor, plays an important role in regulating cell functions such as proliferation and differentiation. Most proteins are unstable, and protein stability is regulated through the ubiquitin-proteasome system (UPS) or the autophagy lysosome pathway (ALP). Whether SRF is degraded and what mechanisms control SRF protein stability remain unexplored. Western blot analyses of cells treated with cycloheximide (CHX), a protein synthesis inhibitor, showed that SRF was degraded in a time-dependent manner. Moreover, we observed that SRF undergoes autophagy-dependent destruction, which is accelerated by serum deprivation. Through bioinformatics screening, we found that SRF contains the GSK3ß phosphorylation motif (T/SPPXS): SPDSPPRSDPT, which is conserved from zebrafish to humans. Serum deprivation stimulated GSK3ß activation that then potentiates SRF degradation through the autophagy lysosome pathway. Since SRF is important for numerous cellular activities, our results suggest that the autophagy-dependent SRF degradation pathway may provide a new avenue to modulate SRF-mediated cell functions.


Subject(s)
Autophagy , Serum Response Factor/chemistry , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Culture Media, Serum-Free/pharmacology , Cycloheximide/pharmacology , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Lysosomes/metabolism , Protein Stability , Rats , Serum Response Factor/metabolism
4.
Bioorg Med Chem Lett ; 27(4): 1055-1061, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28089347

ABSTRACT

Retinoid X receptor alpha (RXRα), an important ligand-dependent transcription factor, plays a critical role in the development of various cancers and metabolic and neurodegenerative diseases. Therefore, RXRα represents one of the most important targets in modern drug discovery. In this study, Drugbank 2.0 with 1280 old drugs were virtually screened by Glide according to the crystal structure of ligand-binding domain (LBP) of RXRα. 15 compounds selected were tested for their binding and transcriptional activity toward RXRα by Biacore and reporter gene assay, respectively. The identified new scafford ligand of RXRα, Pitavastatin (1), was chemically optimized. Our results demonstrated that statin compounds Pitavastatin (1) and Fluvastatin (4) could bind to the LBP of RXRα (KD=13.30µM and 11.04µM, respectively) and serve as transcriptional antagonists of RXRα. On the contrary, compound (12) (domperidone) and (13) (rosiglitazone maleate) could bind to the LBP of RXRα (KD=8.80µM and 15.01µM, respectively) but serve as transcriptional agonists of RXRα.


Subject(s)
Databases, Factual , Retinoid X Receptor alpha/antagonists & inhibitors , Crystallography, X-Ray , Drug Evaluation, Preclinical , Fatty Acids, Monounsaturated/chemistry , Fatty Acids, Monounsaturated/pharmacology , Fluvastatin , Indoles/chemistry , Indoles/pharmacology , Ligands , Quinolines/chemistry , Quinolines/pharmacology , Retinoid X Receptor alpha/chemistry
5.
FASEB J ; 29(11): 4523-31, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26183772

ABSTRACT

Coxsackievirus B3 (CVB3), an important human causative pathogen for viral myocarditis, pancreatitis, and meningitis, has evolved different strategies to manipulate the host signaling machinery to ensure successful viral infection. We previously revealed a crucial role for the ERK1/2 signaling pathway in regulating viral infectivity. However, the detail mechanism remains largely unknown. Grb2-associated binder 1 (GAB1) is an important docking protein responsible for intracellular signaling assembly and transduction. In this study, we demonstrated that GAB1 was proteolytically cleaved after CVB3 infection at G175 and G436 by virus-encoded protease 2A(pro), independent of caspase activation. Knockdown of GAB1 resulted in a significant reduction of viral protein expression and virus titers. Moreover, we showed that virus-induced cleavage of GAB1 is beneficial to viral growth as the N-terminal proteolytic product of GAB1 (GAB1-N1-174) further enhances ERK1/2 activation and promotes viral replication. Our results collectively suggest that CVB3 targets host GAB1 to generate a GAB1-N1-174 fragment that enhances viral infectivity, at least in part, via activation of the ERK pathway. The findings in this study suggest a novel mechanism that CVB3 employs to subvert the host signaling and facilitate consequent viral replication.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Enterovirus B, Human/physiology , MAP Kinase Signaling System , Peptide Hydrolases/metabolism , Viral Proteins/metabolism , Virus Replication/physiology , Adaptor Proteins, Signal Transducing/genetics , Caspases/genetics , Caspases/metabolism , Coxsackievirus Infections/genetics , Coxsackievirus Infections/metabolism , Enzyme Activation/genetics , HeLa Cells , Humans , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Peptide Hydrolases/genetics , Viral Proteins/genetics
6.
Cell Prolif ; : e13722, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39072821

ABSTRACT

Aberrant activation of dermal fibroblasts during wound healing often leads to debilitating fibrotic changes in the skin, such as scleroderma and keloids. However, the underlying cellular and molecular mechanisms remain elusive. Here, we established a wound-induced skin fibrosis (WISF) mouse model in mature adult mice, characterised by excessive deposition of collagen bundles, loss of dermal adipocytes, and enrichment of DPP4+Ly6A+THY1+ hypodermal interstitial adipocyte progenitors (HI-APs) and pericytes, resembling human fibrotic skin diseases. This WISF model exhibited an age-dependent gain of fibrotic characteristics, contrasting with the wound-induced hair neogenesis observed in younger mice. Through comprehensive analyses of the WISF, we delineated a trajectory of fibroblast differentiation that originates from HI-APs. These progenitors highly expressed several extracellular matrix (ECM) genes and exhibited a TGFß pathway signature. TGFß was identified as the key signal to inhibit the adipogenic potential and maintain the fibrogenic potential of dermal APs. Additionally, administering a TGFß receptor inhibitor to wound scar reduced the abundance of ECM-producing APs. Finally, analysis of human scleroderma skin tissues revealed a negative correlation between the expression of AP-, ECM-, and TGFß pathway-related genes and PPARG. Overall, this study establishes a wound-induced skin fibrosis mouse model and demonstrates that TGFß-mediated blockage of HI-AP differentiation is crucial for driving fibrotic pathology. Targeting HI-APs and adipogenesis may provide novel avenues for developing disease-modifying therapies for fibrotic skin diseases.

7.
Elife ; 132024 Aug 30.
Article in English | MEDLINE | ID: mdl-39213029

ABSTRACT

Allergic contact dermatitis (ACD), a prevalent inflammatory skin disease, is elicited upon repeated skin contact with protein-reactive chemicals through a complex and poorly characterized cellular network between immune cells and skin resident cells. Here, single-cell transcriptomic analysis of the murine hapten-elicited model of ACD reveals that upon elicitation of ACD, infiltrated CD4+ or CD8+ lymphocytes were primarily the IFNγ-producing type 1 central memory phenotype. In contrast, type 2 cytokines (IL4 and IL13) were dominantly expressed by basophils, IL17A was primarily expressed by δγ T cells, and IL1ß was identified as the primary cytokine expressed by activated neutrophils/monocytes and macrophages. Furthermore, analysis of skin resident cells identified a sub-cluster of dermal fibroblasts with preadipocyte signature as a prominent target for IFNγ+ lymphocytes and dermal source for key T cell chemokines CXCL9/10. IFNγ treatment shifted dermal fibroblasts from collagen-producing to CXCL9/10-producing, which promoted T cell polarization toward the type-1 phenotype through a CXCR3-dependent mechanism. Furthermore, targeted deletion of Ifngr1 in dermal fibroblasts in mice reduced Cxcl9/10 expression, dermal infiltration of CD8+ T cell, and alleviated ACD inflammation in mice. Finally, we showed that IFNγ+ CD8+ T cells and CXCL10-producing dermal fibroblasts co-enriched in the dermis of human ACD skin. Together, our results define the cell type-specific immune responses in ACD, and recognize an indispensable role of dermal fibroblasts in shaping the development of type-1 skin inflammation through the IFNGR-CXCR3 signaling circuit during ACD pathogenesis.


Subject(s)
Dermatitis, Allergic Contact , Disease Models, Animal , Animals , Mice , Dermatitis, Allergic Contact/immunology , Dermatitis, Allergic Contact/genetics , Fibroblasts/metabolism , Fibroblasts/immunology , Single-Cell Analysis , Transcriptome , Gene Expression Profiling , Mice, Inbred C57BL , Receptors, CXCR3/genetics , Receptors, CXCR3/metabolism , Interferon-gamma/metabolism , CD8-Positive T-Lymphocytes/immunology , Skin/immunology , Skin/pathology , Receptors, Interferon/genetics , Receptors, Interferon/metabolism , Cytokines/metabolism , Female , Interferon gamma Receptor
8.
bioRxiv ; 2024 Jan 29.
Article in English | MEDLINE | ID: mdl-38352498

ABSTRACT

Cancer development and progression are generally associated with dysregulation of gene expression, often resulting from changes in transcription factor (TF) sequence or expression. Identifying key TFs involved in cancer gene regulation provides a framework for potential new therapeutics. This study presents a large-scale cancer gene TF-DNA interaction network as well as an extensive promoter clone resource for future studies. Most highly connected TFs do not show a preference for binding to promoters of genes associated with either good or poor cancer prognosis, suggesting that emerging strategies aimed at shifting gene expression balance between these two prognostic groups may be inherently complex. However, we identified potential for oncogene targeted therapeutics, with half of the tested oncogenes being potentially repressed by influencing specific activator or bifunctional TFs. Finally, we investigate the role of intrinsically disordered regions within the key cancer-related TF estrogen receptor ɑ (ESR1) on DNA binding and transcriptional activity, and found that these regions can have complex trade-offs in TF function. Altogether, our study not only broadens our knowledge of TFs involved in the cancer gene regulatory network but also provides a valuable resource for future studies, laying a foundation for potential therapeutic strategies targeting TFs in cancer.

9.
Life Sci Alliance ; 7(10)2024 Oct.
Article in English | MEDLINE | ID: mdl-39013578

ABSTRACT

Cancer development and progression are generally associated with gene dysregulation, often resulting from changes in the transcription factor (TF) sequence or expression. Identifying key TFs involved in cancer gene regulation provides a framework for potential new therapeutics. This study presents a large-scale cancer gene TF-DNA interaction network, as well as an extensive promoter clone resource for future studies. Highly connected TFs bind to promoters of genes associated with either good or poor cancer prognosis, suggesting that strategies aimed at shifting gene expression balance between these two prognostic groups may be inherently complex. However, we identified potential for oncogene-targeted therapeutics, with half of the tested oncogenes being potentially repressed by influencing specific activators or bifunctional TFs. Finally, we investigate the role of intrinsically disordered regions within the key cancer-related TF ESR1 in DNA binding and transcriptional activity, and found that these regions can have complex trade-offs in TF function. Altogether, our study broadens our knowledge of the TFs involved in cancer gene regulation and provides a valuable resource for future studies and therapeutics.


Subject(s)
DNA , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Neoplasms , Protein Binding , Transcription Factors , Humans , Neoplasms/genetics , Neoplasms/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , DNA/metabolism , DNA/genetics , Promoter Regions, Genetic/genetics , Oncogenes/genetics , Prognosis , Estrogen Receptor alpha/metabolism , Estrogen Receptor alpha/genetics , Computational Biology/methods
10.
Nat Commun ; 14(1): 913, 2023 02 17.
Article in English | MEDLINE | ID: mdl-36808133

ABSTRACT

Although >90% of somatic mutations reside in non-coding regions, few have been reported as cancer drivers. To predict driver non-coding variants (NCVs), we present a transcription factor (TF)-aware burden test based on a model of coherent TF function in promoters. We apply this test to NCVs from the Pan-Cancer Analysis of Whole Genomes cohort and predict 2555 driver NCVs in the promoters of 813 genes across 20 cancer types. These genes are enriched in cancer-related gene ontologies, essential genes, and genes associated with cancer prognosis. We find that 765 candidate driver NCVs alter transcriptional activity, 510 lead to differential binding of TF-cofactor regulatory complexes, and that they primarily impact the binding of ETS factors. Finally, we show that different NCVs within a promoter often affect transcriptional activity through shared mechanisms. Our integrated computational and experimental approach shows that cancer NCVs are widespread and that ETS factors are commonly disrupted.


Subject(s)
Neoplasms , Humans , Mutation , Neoplasms/genetics , Binding Sites/genetics , Transcription Factors/metabolism , Gene Expression Regulation
11.
Cell Rep ; 42(6): 112647, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37330908

ABSTRACT

Dermal adipocyte lineage cells are highly plastic and can undergo reversible differentiation and dedifferentiation in response to various stimuli. Using single-cell RNA sequencing of developing or wounded mouse skin, we classify dermal fibroblasts (dFBs) into distinct non-adipogenic and adipogenic cell states. Cell differentiation trajectory analyses identify IL-1-NF-κB and WNT-ß-catenin as top signaling pathways that positively and negatively associate with adipogenesis, respectively. Upon wounding, activation of adipocyte progenitors and wound-induced adipogenesis are mediated in part by neutrophils through the IL-1R-NF-κB-CREB signaling axis. In contrast, WNT activation, by WNT ligand and/or ablation of Gsk3, inhibits the adipogenic potential of dFBs but promotes lipolysis and dedifferentiation of mature adipocytes, contributing to myofibroblast formation. Finally, sustained WNT activation and inhibition of adipogenesis is seen in human keloids. These data reveal molecular mechanisms underlying the plasticity of dermal adipocyte lineage cells, defining potential therapeutic targets for defective wound healing and scar formation.


Subject(s)
Glycogen Synthase Kinase 3 , NF-kappa B , Mice , Animals , Humans , NF-kappa B/metabolism , Glycogen Synthase Kinase 3/metabolism , Cell Differentiation/physiology , Adipocytes/metabolism , Wnt Signaling Pathway/physiology , Adipogenesis/genetics , Interleukin-1/metabolism , beta Catenin/metabolism
12.
Am J Physiol Heart Circ Physiol ; 301(2): H459-68, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21572014

ABSTRACT

Metformin is the first choice drug for the treatment of patients with diabetes, but its use is debated in patients with advanced cardiorenal disease. Epidemiological data suggest that metformin may reduce cardiac events, in patients both with and without heart failure. Experimental evidence suggests that metformin reduces cardiac ischemia-reperfusion injury. It is unknown whether metformin improves cardiac function (remodeling) in a long-term post-MI remodeling model. We therefore studied male, nondiabetic, Sprague-Dawley rats that were subjected to either myocardial infarction (MI) or sham operation. Animals were randomly allocated to treatment with normal water or metformin-containing water (250 mg·kg(-1)·day(-1)). At baseline, 6 wk, and 12 wk, metabolic parameters were analyzed and oral glucose tolerance tests (OGTT) were performed. Echocardiography and hemodynamic parameters were assessed 12 wk after MI. In the MI model, infarct size was significantly smaller after 12-wk metformin treatment (29.6 ± 3.2 vs. 38.0 ± 2.2%, P < 0.05). Moreover, metformin resulted in less left ventricular dilatation (6.0 ± 0.4 vs. 7.6 ± 0.6 mm, P < 0.05) and preservation of left ventricular ejection fraction (65.8 ± 3.7% vs. 48.6 ± 5.6%, P < 0.05) compared with MI control. The improved cardiac function was associated with decreased atrial natriuretic peptide mRNA levels in the metformin-treated group (50% reduction compared with MI, P < 0.05). Insulin resistance did not occur during cardiac remodeling (as indicated by normal OGTT) and fasting glucose levels and the pattern of the OGTT were not affected by metformin. Molecular analyses suggested that altered AMP kinase phosphorylation status and low insulin levels mediate the salutary effects of metformin. Altogether our results indicate that metformin may have potential to attenuate heart failure development after myocardial infarction, in the absence of diabetes and independent of systemic glucose levels.


Subject(s)
Cardiotonic Agents/pharmacology , Heart Failure/drug therapy , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Myocardial Infarction/drug therapy , Ventricular Function, Left/drug effects , AMP-Activated Protein Kinases/metabolism , Animals , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Blood Glucose/drug effects , Disease Models, Animal , Energy Metabolism/drug effects , Fibrosis , Gene Expression Regulation/drug effects , Glucose Tolerance Test , Heart Failure/blood , Heart Failure/etiology , Heart Failure/pathology , Heart Failure/physiopathology , Hypertrophy, Left Ventricular/etiology , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Insulin/blood , Male , Myocardial Infarction/blood , Myocardial Infarction/complications , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/metabolism , Myocardium/pathology , Phosphorylation , Rats , Rats, Sprague-Dawley , Stroke Volume/drug effects , Time Factors , Ventricular Remodeling/drug effects
13.
Cardiovasc Diabetol ; 10: 85, 2011 Sep 28.
Article in English | MEDLINE | ID: mdl-21955567

ABSTRACT

BACKGROUND: Progressive remodeling after myocardial infarction (MI) is a leading cause of morbidity and mortality. Recently, glucagon-like peptide (GLP)-1 was shown to have cardioprotective effects, but treatment with GLP-1 is limited by its short half-life. It is rapidly degraded by the enzyme dipeptidyl peptidase-4 (DPP-4), an enzyme which inhibits GLP-1 activity. We hypothesized that the DPP-4 inhibitor vildagliptin will increase levels of GLP-1 and may exert protective effects on cardiac function after MI. METHODS: Sprague-Dawley rats were either subjected to coronary ligation to induce MI and left ventricular (LV) remodeling, or sham operation. Parts of the rats with an MI were pre-treated for 2 days with the DPP-4 inhibitor vildagliptin (MI-Vildagliptin immediate, MI-VI, 15 mg/kg/day). The remainder of the rats was, three weeks after coronary artery ligation, subjected to treatment with DPP-4 inhibitor vildagliptin (MI-Vildagliptin Late, MI-VL) or control (MI). At 12 weeks, echocardiography and invasive hemodynamics were measured and molecular analysis and immunohistochemistry were performed. RESULTS: Vildagliptin inhibited the DPP-4 enzymatic activity by almost 70% and increased active GLP-1 levels by about 3-fold in plasma in both treated groups (p < 0.05 vs. non-treated groups). Cardiac function (ejection fraction) was decreased in all 3 MI groups compared with Sham group (p < 0.05); treatment with vildagliptin, either early or late, did not reverse cardiac remodeling. ANP (atrial natriuretic peptide) and BNP (brain natriuretic peptide) mRNA levels were significantly increased in all 3 MI groups, but no significant reductions were observed in both vildagliptin groups. Vildagliptin also did not change cardiomyocyte size or capillary density after MI. No effects were detected on glucose level and body weight in the post-MI remodeling model. CONCLUSION: Vildagliptin increases the active GLP-1 level via inhibition of DPP-4, but it has no substantial protective effects on cardiac function in this well established long-term post-MI cardiac remodeling model.


Subject(s)
Adamantane/analogs & derivatives , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Disease Models, Animal , Heart Failure/drug therapy , Myocardial Infarction/drug therapy , Nitriles/administration & dosage , Pyrrolidines/administration & dosage , Adamantane/administration & dosage , Animals , Dipeptidyl Peptidase 4/metabolism , Drug Administration Schedule , Heart Failure/enzymology , Male , Myocardial Infarction/enzymology , Random Allocation , Rats , Rats, Sprague-Dawley , Time Factors , Vildagliptin
14.
Sci Transl Med ; 13(577)2021 01 20.
Article in English | MEDLINE | ID: mdl-33472955

ABSTRACT

Infections are a major complication of obesity, but the mechanisms responsible for impaired defense against microbes are not well understood. Here, we found that adipocyte progenitors were lost from the dermis during diet-induced obesity (DIO) in humans and mice. The loss of adipogenic fibroblasts from mice resulted in less antimicrobial peptide production and greatly increased susceptibility to Staphylococcus aureus infection. The decrease in adipocyte progenitors in DIO mice was explained by expression of transforming growth factor-ß (TGFß) by mature adipocytes that then inhibited adipocyte progenitors and the production of cathelicidin in vitro. Administration of a TGFß receptor inhibitor or a peroxisome proliferator-activated receptor-γ agonist reversed this inhibition in both cultured adipocyte progenitors and in mice and subsequently restored the capacity of obese mice to defend against S. aureus skin infection. Together, these results explain how obesity promotes dysfunction of the antimicrobial function of reactive dermal adipogenesis and identifies potential therapeutic targets to manage skin infection associated with obesity.


Subject(s)
Adipocytes/immunology , Anti-Infective Agents , Obesity/complications , Staphylococcal Infections/immunology , 3T3-L1 Cells , Adipocytes/microbiology , Animals , Anti-Infective Agents/pharmacology , Cell Differentiation , Diet , Diet, High-Fat , Mice , Mice, Inbred C57BL , PPAR gamma/agonists , Staphylococcal Infections/complications , Staphylococcus aureus , Transforming Growth Factor beta/antagonists & inhibitors
15.
Cells ; 8(8)2019 08 01.
Article in English | MEDLINE | ID: mdl-31374826

ABSTRACT

Located at the skin surface, keratinocytes (KCs) are constantly exposed to external stimuli and are the first responders to invading pathogens and injury. Upon skin injury, activated KCs secrete an array of alarmin molecules, providing a rapid and specific innate immune response against danger signals. However, dysregulation of the innate immune response of KCs may lead to uncontrolled inflammation and psoriasis pathogenesis. Keratins (KRT) are the major structural intermediate filament proteins in KCs and are expressed in a highly specific pattern at different differentiation stages of KCs. While KRT14-KRT5 is restricted to basal proliferative KCs, and KRT10-KRT1 is restricted to suprabasal differentiated KCs in normal skin epidermis, the wound proximal KCs downregulate KRT10-K1 and upregulate KRT16/KRT17-KRT6 upon skin injury. Recent studies have recognized KRT6/16/17 as key early barrier alarmins and upregulation of these keratins alters proliferation, cell adhesion, migration and inflammatory features of KCs, contributing to hyperproliferation and innate immune activation of KCs in response to an epidermal barrier breach, followed by the autoimmune activation of T cells that drives psoriasis. Here, we have reviewed how keratins are dysregulated during skin injury, their roles in wound repairs and in initiating the innate immune system and the subsequent autoimmune amplification that arises in psoriasis.


Subject(s)
Keratin-16/metabolism , Keratin-17/metabolism , Keratin-6/metabolism , Psoriasis/metabolism , Wound Healing , Alarmins/metabolism , Cell Adhesion , Cell Differentiation , Cell Movement , Cell Proliferation , Gene Expression Regulation , Humans , Immunity, Innate
16.
Mol Cancer Ther ; 18(5): 886-899, 2019 05.
Article in English | MEDLINE | ID: mdl-30926635

ABSTRACT

Nur77 (also called TR3 or NGFI-B), an orphan member of the nuclear receptor superfamily, induces apoptosis by translocating to mitochondria where it interacts with Bcl-2 to convert Bcl-2 from an antiapoptotic to a pro-apoptotic molecule. Nur77 posttranslational modification such as phosphorylation has been shown to induce Nur77 translocation from the nucleus to mitochondria. However, small molecules that can bind directly to Nur77 to trigger its mitochondrial localization and Bcl-2 interaction remain to be explored. Here, we report our identification and characterization of DIM-C-pPhCF3 +MeSO3 - (BI1071), an oxidized product derived from indole-3-carbinol metabolite, as a modulator of the Nur77-Bcl-2 apoptotic pathway. BI1071 binds Nur77 with high affinity, promotes Nur77 mitochondrial targeting and interaction with Bcl-2, and effectively induces apoptosis of cancer cells in a Nur77- and Bcl-2-dependent manner. Studies with animal model showed that BI1071 potently inhibited the growth of tumor cells in animals through its induction of apoptosis. Our results identify BI1071 as a novel Nur77-binding modulator of the Nur77-Bcl-2 apoptotic pathway, which may serve as a promising lead for treating cancers with overexpression of Bcl-2.


Subject(s)
Hydrocarbons, Fluorinated/pharmacology , Indoles/pharmacology , Neoplasms/drug therapy , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Animals , Apoptosis/drug effects , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , Indoles/chemistry , MCF-7 Cells , Mice , Mitochondria/drug effects , Mitochondria/genetics , Neoplasms/genetics , Neoplasms/pathology , Protein Binding , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
17.
Theranostics ; 8(11): 3007-3021, 2018.
Article in English | MEDLINE | ID: mdl-29896299

ABSTRACT

Rationale: Atherosclerosis is a chronic inflammatory and epigenetic disease that is influenced by different patterns of blood flow. However, the epigenetic mechanism whereby atheroprotective flow controls endothelial gene programming remains elusive. Here, we investigated the possibility that flow alters endothelial gene expression through epigenetic mechanisms. Methods: En face staining and western blot were used to detect protein expression. Real-time PCR was used to determine relative gene expression. RNA-sequencing of human umbilical vein endothelial cells treated with siRNA of enhancer of zeste homolog 2 (EZH2) or laminar flow was used for transcriptional profiling. Results: We found that trimethylation of histone 3 lysine 27 (H3K27me3), a repressive epigenetic mark that orchestrates gene repression, was reduced in laminar flow areas of mouse aorta and flow-treated human endothelial cells. The decrease of H3K27me3 paralleled a reduction in the epigenetic "writer"-EZH2, the catalytic subunit of the polycomb repressive complex 2 (PRC2). Moreover, laminar flow decreased expression of EZH2 via mechanosensitive miR101. Genome-wide transcriptome profiling studies in endothelial cells treated with EZH2 siRNA and flow revealed the upregulation of novel mechanosensitive gene IGFBP5 (insulin-like growth factor-binding protein 5), which is epigenetically silenced by H3K27me3. Functionally, inhibition of H3K27me3 by EZH2 siRNA or GSK126 (a specific EZH2 inhibitor) reduced H3K27me3 levels and monocyte adhesion to endothelial cells. Adenoviral overexpression of IGFBP5 also recapitulated the anti-inflammatory effects of H3K27me3 inhibition. More importantly, we observed EZH2 upregulation, and IGFBP5 downregulation, in advanced atherosclerotic plaques from human patients. Conclusion: Taken together, our findings reveal that atheroprotective flow reduces H3K27me3 as a chromatin-based mechanism to augment the expression of genes that confer an anti-inflammatory response in the endothelium. Our study exemplifies flow-dependent epigenetic regulation of endothelial gene expression, and also suggests that targeting the EZH2/H3K27me3/IGFBP5 pathway may offer novel therapeutics for inflammatory disorders such as atherosclerosis.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Atherosclerosis/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic/genetics , Histones/genetics , Insulin-Like Growth Factor Binding Protein 5/genetics , Animals , Atherosclerosis/immunology , Atherosclerosis/therapy , Endothelium/immunology , Genetic Therapy , Human Umbilical Vein Endothelial Cells/immunology , Humans , Methylation , Mice , Polycomb Repressive Complex 2/genetics , RNA, Small Interfering/genetics
18.
Transl Res ; 176: 18-28.e2, 2016 10.
Article in English | MEDLINE | ID: mdl-27295628

ABSTRACT

Atherosclerosis is a mechanobiology-related disease that preferentially develops in the aortic arch and arterial branches, which are exposed to disturbed/turbulent blood flow but less in thoracic aorta where the flow pattern is steady laminar flow (LF). Increasing evidence supports that steady LF with high shear stress is protective against atherosclerosis. However, the molecular mechanisms of LF-mediated atheroprotection remain incompletely understood. Hippo/YAP (yes-associated protein) pathway senses and effects mechanical cues and has been reported to be a master regulator of cell proliferation, differentiation, and tissue homeostasis. Here, we show that LF inhibits YAP activity in endothelial cells (ECs). We observed that YAP is highly expressed in mouse EC-enriched tissues (lung and aorta) and in human ECs. Furthermore, we found in apolipoprotein E deficient (ApoE(-/-)) mice and human ECs, LF decreased the level of nuclear YAP protein and YAP target gene expression (connective tissue growth factor and cysteine-rich protein 61) through promoting Hippo kinases LATS1/2-dependent YAP (Serine 127) phosphorylation. Functionally, we revealed that YAP depletion in ECs phenocopying LF responses, reduced the expression of cell cycle gene cyclin A1 (CCNA1) and proinflammatory gene CCL2 (MCP-1). Taken together, we demonstrate that atheroprotective LF inhibits endothelial YAP activation, which may contribute to LF-mediated ECs quiescence and anti-inflammation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Atherosclerosis/metabolism , Atherosclerosis/pathology , Coronary Circulation , Endothelial Cells/metabolism , Phosphoproteins/metabolism , Signal Transduction , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Cell Cycle Proteins , Cell Nucleus/metabolism , Chemokine CCL2/metabolism , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Cyclin A1/metabolism , Cysteine-Rich Protein 61/genetics , Cysteine-Rich Protein 61/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Mice, Inbred C57BL , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein Transport , Transcription Factors , Tumor Suppressor Proteins/metabolism , YAP-Signaling Proteins
19.
Oncotarget ; 7(25): 37622-37635, 2016 Jun 21.
Article in English | MEDLINE | ID: mdl-27191895

ABSTRACT

TRPV4 ion channel mediates vascular mechanosensitivity and vasodilation. Here, we sought to explore whether non-mechanical activation of TRPV4 could limit vascular inflammation and atherosclerosis. We found that GSK1016790A, a potent and specific small-molecule agonist of TRPV4, induces the phosphorylation and activation of eNOS partially through the AMPK pathway. Moreover, GSK1016790A inhibited TNF-α-induced monocyte adhesion to human endothelial cells. Mice given GSK1016790A showed increased phosphorylation of eNOS and AMPK in the aorta and decreased leukocyte adhesion to TNF-α-inflamed endothelium. Importantly, oral administration of GSK1016790A reduced atherosclerotic plaque formation in ApoE deficient mice fed a Western-type diet. Together, the present study suggests that pharmacological activation of TRPV4 may serve as a potential therapeutic approach to treat atherosclerosis.


Subject(s)
Atherosclerosis/drug therapy , Monocytes/drug effects , TRPV Cation Channels/agonists , Animals , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cell Adhesion/drug effects , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells , Humans , Leucine/analogs & derivatives , Leucine/pharmacology , Male , Mice , Mice, Inbred C57BL , Monocytes/pathology , Sulfonamides/pharmacology , TRPV Cation Channels/genetics , U937 Cells
20.
Aging (Albany NY) ; 8(5): 1064-82, 2016 05.
Article in English | MEDLINE | ID: mdl-27249230

ABSTRACT

SIRT6 is an important member of sirtuin family that represses inflammation, aging and DNA damage, three of which are causing factors for endothelial dysfunction. SIRT6 expression is decreased in atherosclerotic lesions from ApoE(-/-) mice and human patients. However, the role of SIRT6 in regulating vascular endothelial function and atherosclerosis is not well understood. Here we show that SIRT6 protects against endothelial dysfunction and atherosclerosis. Global and endothelium-specific SIRT6 knockout mice exhibited impaired endothelium-dependent vasorelaxation. Moreover, SIRT6(+/-) haploinsufficient mice fed a high-fat diet (HFD) also displayed impaired endothelium-dependent vasorelaxation. Importantly, SIRT6(+/-); ApoE(-/-) mice after HFD feeding exhibited exacerbated atherosclerotic lesion development, concurrent with increased expression of the proinflammatory cytokine VCAM-1. Loss- and gain-of-SIRT6 function studies in cultured human endothelial cells (ECs) showed that SIRT6 attenuated monocyte adhesion to ECs. RNA-sequencing profiling revealed that SIRT6 overexpression decreased the expression of multiple atherosclerosis-related genes, including proatherogenic gene TNFSF4 (tumor necrosis factor superfamily member 4). Chromatin immunoprecipitation assays showed that SIRT6 decreased TNFSF4 gene expression by binding to and deacetylating H3K9 at TNFSF4 gene promoter. Collectively, these findings demonstrate that SIRT6 play a pivotal role in maintaining endothelial function and increased SIRT6 activity could be a new therapeutic strategy to combat atherosclerotic disease.


Subject(s)
Atherosclerosis/metabolism , Endothelium, Vascular/metabolism , Sirtuins/metabolism , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Atherosclerosis/genetics , Cell Adhesion/physiology , Cells, Cultured , Endothelial Cells/metabolism , Haploinsufficiency , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , OX40 Ligand , Sirtuins/genetics , Tumor Necrosis Factors/genetics , Tumor Necrosis Factors/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism , Vasodilation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL