Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
J Immunol ; 203(11): 3045-3053, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31611260

ABSTRACT

Macrophages drive the pathological process of inflammatory bowel diseases (IBD) mostly by secreting proinflammatory cytokines, such as Tnf-α. Recent studies have indicated the association between epigenetic modifications and macrophage functions. However, epigenetic mechanisms regulating macrophages' functional involvement in IBD remain unknown. In this study, we investigated whether the epigenetic regulator Uhrf1 plays a role in innate immunity by functionally regulating macrophages in intestines. We employed two transgenic strains of mice (one with Uhrf1 deficiency in macrophages [Uhrf1fl/flLyz2-Cre mice] and the other with the two mutations at Uhrf1's DNA methylation regulatory site [Uhrf1YP187/188AA mice]) to assess their susceptibility to dextran sodium sulfate-induced colitis. We examined the cytokines derived from Uhrf1fl/flLyz2-Cre and Uhrf1YP187/188AA macrophages in response to LPS stimulation. We also analyzed the effects of proinflammatory cytokines on Uhrf1 expression in macrophages. The data demonstrated that Uhrf1 deficiency and Uhrf1YP187/188AA mutation resulted in severe colitis in the dextran sodium sulfate-treated mice. In vitro analysis revealed the hypomethylation of Tnf-α promoter and the increased Tnf-α expression in Uhrf1fl/flLyz2-Cre and Uhrf1YP187/188AA macrophages in response to LPS stimulation, and anti-Tnf-α therapy implied the key role of Tnf-α to the aggravated colitis in Uhrf1-deficient mice. Exogenous Tnf-α destabilized Uhrf1 protein through ubiquitination-mediated protein degradation, triggering macrophage activation. In conclusion, we identified that Uhrf1-mediated DNA methylation controls Tnf-α expression of macrophages in the experimental colitis resembling IBD. The epigenetic mechanisms that activate macrophages may provide new therapeutic targets for IBD treatment.


Subject(s)
CCAAT-Enhancer-Binding Proteins/immunology , Colitis/immunology , Inflammation/immunology , Inflammatory Bowel Diseases/immunology , Macrophages/immunology , Tumor Necrosis Factor-alpha/immunology , Ubiquitin-Protein Ligases/immunology , Animals , CCAAT-Enhancer-Binding Proteins/deficiency , CCAAT-Enhancer-Binding Proteins/genetics , Disease Models, Animal , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Methylation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Tumor Necrosis Factor-alpha/genetics , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics
2.
FASEB J ; 33(7): 8294-8305, 2019 07.
Article in English | MEDLINE | ID: mdl-30995416

ABSTRACT

The ubiquitin-like, containing PHD and RING finger domains, 1 (UHRF1) protein recognizes DNA methylation and histone modification and plays a critical role in epigenetic regulation. Recently, UHRF1 was shown to have a role in DNA methylation in oocytes and early embryos. Here, we reveal that maternal UHRF1 determines the quality of mouse oocytes. We generated oocyte-specific Uhrf1-knockout mice and found that females were sterile, and few maternal UHRF1-null embryos developed into blastocysts. The UHRF1-null oocytes had an increased incidence of aneuploidy and DNA damage. In addition to defective DNA methylation, histone modification was affected during oogenesis, with UHRF1-null germinal vesicle and metaphase II-stage oocytes exhibiting reduced global histone H3 lysine 9 dimethylation levels and elevated acetylation of histone H4 lysine 12. Taken together, our results suggest that UHRF1 plays an important role in determining oocyte quality and affects epigenetic regulation of oocyte maturation as a maternal protein, which is crucial for embryo developmental potential. Further exploration of the biologic function and underlying mechanisms of maternal UHRF1 will enhance our understanding of the maternal control of the oocyte and early embryonic development.-Cao, Y., Li, M., Liu, F., Ni, X., Wang, S., Zhang, H., Sui, X., Huo, R. Deletion of maternal UHRF1 severely reduces mouse oocyte quality and causes developmental defects in preimplantation embryos.


Subject(s)
Blastocyst/metabolism , CCAAT-Enhancer-Binding Proteins/deficiency , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Oocytes/metabolism , Ubiquitin-Protein Ligases/deficiency , Animals , Blastocyst/pathology , CCAAT-Enhancer-Binding Proteins/metabolism , DNA Damage , DNA Methylation/genetics , Female , Metaphase , Mice , Mice, Knockout , Oocytes/pathology , Ubiquitin-Protein Ligases/metabolism
3.
Prostate ; 79(3): 302-311, 2019 02.
Article in English | MEDLINE | ID: mdl-30430607

ABSTRACT

BACKGROUND: The transcription factor CCAAT-enhancer-binding protein alpha (CEBPA) is a crucial regulator of cell proliferation and differentiation. Expression levels of CEBPA have been suggested to be prognostic in various tumor types. METHODS: Here, we analyzed the immunohistochemical expression of CEBPA in a tissue microarray containing more than 17 000 prostate cancer specimens with annotated clinical and molecular data including for example TMPRSS2:ERG fusion and PTEN deletion status. RESULTS: Normal prostate glands showed moderate to strong CEBPA staining, while CEBPA expression was frequently reduced (40%) or lost (30%) in prostate cancers. Absence of detectable CEBPA expression was markedly more frequent in ERG negative (45%) as compared to ERG positive cancers (20%, P < 0.0001). Reduced CEBPA expression was linked to unfavorable phenotype (P < 0.0001) and poor prognosis (P = 0.0008). Subgroup analyses revealed, that the prognostic value of CEBPA loss was entirely driven by tumors carrying both TMPRSS2:ERG fusions and PTEN deletions. In this subgroup, CEBPA loss was tightly linked to advanced tumor stage (P < 0.0001), high Gleason grade (P < 0.0001), positive nodal stage (0.0003), and early biochemical recurrence (P = 0.0007), while these associations were absent or markedly diminished in tumors with normal PTEN copy numbers and/or absence of ERG fusion. CONCLUSIONS: CEBPA is down regulated in about one third of prostate cancers, but the clinical impact of CEBPA loss is strictly limited to the subset of about 10% prostate cancers carrying both ERG fusion and deletions of the PTEN tumor suppressor. Our findings challenge the concept that prognostic molecular markers may be generally applicable to all prostate cancers.


Subject(s)
CCAAT-Enhancer-Binding Proteins/deficiency , Oncogene Proteins, Fusion/metabolism , PTEN Phosphohydrolase/deficiency , Prostatic Neoplasms/metabolism , Aged , CCAAT-Enhancer-Binding Proteins/biosynthesis , CCAAT-Enhancer-Binding Proteins/metabolism , Gene Dosage , Humans , Immunohistochemistry , Male , Middle Aged , Oncogene Proteins, Fusion/genetics , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Prognosis , Prostatectomy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Tissue Array Analysis
4.
J Pathol ; 244(3): 271-282, 2018 03.
Article in English | MEDLINE | ID: mdl-29083488

ABSTRACT

Osteoclast lineage commitment and differentiation have been studied extensively, although the mechanism by which transcription factor(s) control osteoclast terminal differentiation, activation, and function remains unclear. CCAAT/enhancer-binding protein α (C/ebpα) has been reported to be a key regulator of osteoclast cell lineage commitment, yet C/ebpα's roles in osteoclast terminal differentiation, activation and function, and bone homeostasis, under physiological or pathological conditions, have not been studied because newborn C/ebpα-null mice die within several hours after birth. Furthermore, the function of C/ebpα in osteoclast terminal differentiation, activation, and function is largely unknown. Herein, we generated and analyzed an osteoclast-specific C/ebpα conditional knockout (CKO) mouse model via Ctsk-Cre mice and found that C/ebpα-deficient mice exhibited a severe osteopetrosis phenotype due to impaired osteoclast terminal differentiation, activation, and function, including mildly reduced osteoclast number, impaired osteoclast polarization, actin formation, and bone resorption, which demonstrated the novel function of C/ebpα in cell function and terminal differentiation. Interestingly, C/ebpα deficiency did not affect bone formation or monocyte/macrophage development. Our results further demonstrated that C/ebpα deficiency suppressed the expression of osteoclast functional genes, e.g. encoding cathepsin K (Ctsk), Atp6i (Tcirg1), and osteoclast regulator genes, e.g. encoding c-fos (Fos), and nuclear factor of activated T-cells 1 (Nfatc1), while having no effect on Pu.1 (Spi1) expression. Promoter activity mapping and ChIP assay defined the critical cis-regulatory element (CCRE) in the promoter region of Nfatc1, and also showed that the CCREs were directly associated with C/ebpα, which enhanced the promoter's activity. The deficiency of C/ebpα in osteoclasts completely blocked ovariectomy-induced bone loss, indicating that C/ebpα is a promising new target for the treatment of osteolytic diseases. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Cell Differentiation , NFATC Transcription Factors/metabolism , Osteoclasts/metabolism , Osteogenesis , Animals , Binding Sites , CCAAT-Enhancer-Binding Proteins/deficiency , CCAAT-Enhancer-Binding Proteins/genetics , Cell Lineage , Cells, Cultured , Disease Models, Animal , Female , Gene Expression Regulation, Developmental , Genetic Predisposition to Disease , Homeostasis , Humans , Male , Mice, Knockout , NFATC Transcription Factors/genetics , Osteoclasts/pathology , Osteopetrosis/genetics , Osteopetrosis/metabolism , Osteopetrosis/pathology , Osteoporosis, Postmenopausal/genetics , Osteoporosis, Postmenopausal/metabolism , Osteoporosis, Postmenopausal/pathology , Ovariectomy , Phenotype , Promoter Regions, Genetic , Signal Transduction
5.
J Pathol ; 238(3): 423-33, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26497117

ABSTRACT

The cellular defence protein Nrf2 is a mediator of oncogenesis in pancreatic ductal adenocarcinoma (PDAC) and other cancers. However, the control of Nrf2 expression and activity in cancer is not fully understood. We previously reported the absence of Keap1, a pivotal regulator of Nrf2, in ∼70% of PDAC cases. Here we describe a novel mechanism whereby the epigenetic regulator UHRF1 suppresses Keap1 protein levels. UHRF1 expression was observed in 20% (5 of 25) of benign pancreatic ducts compared to 86% (114 of 132) of pancreatic tumours, and an inverse relationship between UHRF1 and Keap1 levels in PDAC tumours (n = 124) was apparent (p = 0.002). We also provide evidence that UHRF1-mediated regulation of the Nrf2 pathway contributes to the aggressive behaviour of PDAC. Depletion of UHRF1 from PDAC cells decreased growth and enhanced apoptosis and cell cycle arrest. UHRF1 depletion also led to reduced levels of Nrf2-regulated downstream proteins and was accompanied by heightened oxidative stress, in the form of lower glutathione levels and increased reactive oxygen species. Concomitant depletion of Keap1 and UHRF1 restored Nrf2 levels and reversed cell cycle arrest and the increase in reactive oxygen species. Mechanistically, depletion of UHRF1 reduced global and tumour suppressor promoter methylation in pancreatic cancer cell lines, and KEAP1 gene promoter methylation was reduced in one of three cell lines examined. Thus, methylation of the KEAP1 gene promoter may contribute to the suppression of Keap1 protein levels by UHRF1, although our data suggest that additional mechanisms need to be explored. Finally, we demonstrate that K-Ras drives UHRF1 expression, establishing a novel link between this oncogene and Nrf2-mediated cellular protection. Since UHRF1 over-expression occurs in other cancers, its ability to regulate the Keap1-Nrf2 pathway may be critically important to the malignant behaviour of these cancers.


Subject(s)
CCAAT-Enhancer-Binding Proteins/physiology , Intracellular Signaling Peptides and Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Pancreatic Neoplasms/etiology , CCAAT-Enhancer-Binding Proteins/deficiency , Carcinogenesis , Cell Cycle Checkpoints/physiology , Cell Transformation, Neoplastic/pathology , DNA Methylation/physiology , Humans , Kelch-Like ECH-Associated Protein 1 , Oxidative Stress/physiology , Pancreatic Neoplasms/pathology , Signal Transduction/physiology , Tumor Burden , Tumor Cells, Cultured , Ubiquitin-Protein Ligases
6.
EMBO J ; 31(2): 351-65, 2012 Jan 18.
Article in English | MEDLINE | ID: mdl-22068055

ABSTRACT

The transcription factors that control lineage specification of haematopoietic stem cells (HSCs) have been well described for the myeloid and lymphoid lineages, whereas transcriptional control of erythroid (E) and megakaryocytic (Mk) fate is less understood. We here use conditional removal of the GATA-1 and FOG-1 transcription factors to identify FOG-1 as required for the formation of all committed Mk- and E-lineage progenitors, whereas GATA-1 was observed to be specifically required for E-lineage commitment. FOG-1-deficient HSCs and preMegEs, the latter normally bipotent for the Mk and E lineages, underwent myeloid transcriptional reprogramming, and formed myeloid, but not erythroid and megakaryocytic cells in vitro. These results identify FOG-1 and GATA-1 as required for formation of bipotent Mk/E progenitors and their E-lineage commitment, respectively, and show that FOG-1 mediates transcriptional Mk/E programming of HSCs as well as their subsequent Mk/E-lineage commitment. Finally, C/EBPs and FOG-1 exhibited transcriptional cross-regulation in early myelo-erythroid progenitors making their functional antagonism a potential mechanism for separation of the myeloid and Mk/E lineages.


Subject(s)
Erythropoiesis/genetics , GATA1 Transcription Factor/physiology , Gene Expression Regulation, Developmental/genetics , Megakaryocyte-Erythroid Progenitor Cells/cytology , Nuclear Proteins/physiology , Thrombopoiesis/genetics , Transcription Factors/physiology , Animals , Bone Marrow Cells/cytology , CCAAT-Enhancer-Binding Protein-beta/deficiency , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Proteins/deficiency , CCAAT-Enhancer-Binding Proteins/genetics , Cell Lineage , Cells, Cultured/cytology , Colony-Forming Units Assay , Erythroid Precursor Cells/cytology , GATA1 Transcription Factor/genetics , Megakaryocyte Progenitor Cells/cytology , Mice , Mice, Transgenic , Nuclear Proteins/genetics , Transcription Factors/genetics , Transcription, Genetic
7.
J Gastroenterol Hepatol ; 29(5): 1109-18, 2014 May.
Article in English | MEDLINE | ID: mdl-24329600

ABSTRACT

BACKGROUND AND AIMS: Hepatic steatosis is a metabolic liver disease with the potential to progress to steatohepatitis, cirrhosis, and hepatocellular carcinoma (HCC). The aim of this study was to investigate the impact of CCAAT/enhancer-binding protein homologous protein (CHOP) deficiency in the development of steatosis-associated progression of HCC. METHODS: Eight-week-old wild-type (WT) and CHOP knockout (CHOP-/-) mice were fed a normal or methionine-choline-deficient (MCD) diet. Mice were sacrificed after 3 weeks, and steatosis, inflammation, apoptosis, and liver damage were assessed. We also evaluated fibrosis after 8 weeks of nutrition intervention. To explore the role of CHOP in liver carcinogenesis, 25 mg/kg of diethylnitrosamine (DEN) was injected intraperitoneally into 2-week-old mice, which were then fed the aforementioned diets from 8 to 24 weeks of age. CHOP expression in HCC patient livers was also evaluated. RESULTS: CHOP deficiency did not affect steatosis but significantly reduced apoptotic cells, inflammation scores, and serum liver enzymes. It also significantly suppressed total serum bilirubin levels, fibrotic area size, and messenger RNA expression of profibrotic cytokines. DEN-initiated carcinogenesis was promoted by the MCD diet, while CHOP deficiency significantly attenuated the total number and maximum diameter of tumors and the Ki-67 labeling index. In human livers, CHOP expression was enhanced in parallel with non-alcoholic steatohepatitis-to-HCC progression. CONCLUSIONS: CHOP deficiency attenuated apoptosis, inflammation, fibrosis, and tumorigenesis under fat-loading conditions, indicating that a therapeutic strategy targeting CHOP might be effective for fat-induced liver injury and protecting against promotion of carcinogenesis in patients with liver steatosis.


Subject(s)
CCAAT-Enhancer-Binding Proteins/deficiency , Carcinoma, Hepatocellular/therapy , Choline Deficiency , Fatty Liver/therapy , Liver Cirrhosis/therapy , Liver Neoplasms/therapy , Methionine/deficiency , Aged , Aged, 80 and over , Animals , Carcinoma, Hepatocellular/etiology , Fatty Liver/etiology , Fatty Liver/prevention & control , Female , Humans , Liver Cirrhosis/etiology , Liver Cirrhosis/prevention & control , Liver Neoplasms/etiology , Liver Neoplasms/prevention & control , Male , Mice, Inbred C57BL , Middle Aged , Molecular Targeted Therapy
8.
Blood ; 117(18): 4881-4, 2011 May 05.
Article in English | MEDLINE | ID: mdl-21389317

ABSTRACT

CCAAT/enhancer binding protein-α (CEBPA) mutations in acute myeloid leukemia (AML) patients with a normal karyotype (NK) confer favorable prognosis, whereas NK-AML patients per se are of intermediate risk. This suggests that blocked CEBPA function characterizes NK-AML with favorable outcome. We determined the prognostic significance of CEBPA DNA binding function by enzyme-linked immunosorbent assay in 105 NK-AML patients. Suppressed CEBPA DNA binding was defined by 21 good-risk AML patients with inv(16) or t(8;21) (both abnormalities targeting CEBPA) and 8 NK-AML patients with dominant-negative CEBPA mutations. NK-AML patients with suppressed CEBPA function showed a better overall survival (P = .0231) and disease-free survival (P = .0069) than patients with conserved CEBPA function. Suppressed CEBPA DNA binding was an independent marker for better overall survival and disease-free survival in a multivariable analysis that included FLT3-ITD, NPM1 and CEBPA mutation status, white blood cell count, age and lactate dehydrogenase. These data indicate that suppressed CEBPA function is associated with favorable prognosis in NK-AML patients.


Subject(s)
CCAAT-Enhancer-Binding Proteins/deficiency , CCAAT-Enhancer-Binding Proteins/genetics , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mutation , Adolescent , Adult , Aged , Base Sequence , CCAAT-Enhancer-Binding Proteins/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Female , Humans , Kaplan-Meier Estimate , Karyotyping , Male , Middle Aged , Nucleophosmin , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , RUNX1 Translocation Partner 1 Protein , U937 Cells , Young Adult
10.
Mol Cancer ; 11: 90, 2012 Dec 12.
Article in English | MEDLINE | ID: mdl-23234329

ABSTRACT

BACKGROUND: The CCAAT/enhancer binding proteins (C/EBPs) play important roles in carcinogenesis of many tumors including the lung. Since multiple C/EBPs are expressed in lung, the combinatorial expression of these C/EBPs on lung carcinogenesis is not known. METHODS: A transgenic mouse line expressing a dominant negative A-C/EBP under the promoter of lung epithelial Clara cell secretory protein (CCSP) gene in doxycycline dependent fashion was subjected to 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK)-induced lung carcinogenesis bioassay in the presence and absence of doxycycline, and the effect of abolition of DNA binding activities of C/EBPs on lung carcinogenesis was examined. RESULTS: A-C/EBP expression was found not to interfere with tumor development; however, it suppressed the malignant conversion of adenoma to carcinoma during NNK-induced lung carcinogenesis. The results suggested that Ki67 may be used as a marker for lung carcinomas in mouse. CONCLUSIONS: The DNA binding of C/EBP family members can be used as a potential molecular target for lung cancer therapy.


Subject(s)
Adenoma/genetics , CCAAT-Enhancer-Binding Proteins/deficiency , Carcinoma/genetics , Cell Transformation, Neoplastic/genetics , Lung Neoplasms/genetics , Adenoma/epidemiology , Adenoma/pathology , Animals , Biomarkers, Tumor/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Carcinoma/epidemiology , Carcinoma/pathology , Cell Transformation, Neoplastic/chemically induced , In Situ Nick-End Labeling , Incidence , Ki-67 Antigen/metabolism , Lung Neoplasms/chemically induced , Lung Neoplasms/epidemiology , Lung Neoplasms/pathology , Male , Mice , Mice, Transgenic , Nitrosamines/toxicity , Pulmonary Alveoli/pathology
11.
Circulation ; 122(4): 361-9, 2010 Jul 27.
Article in English | MEDLINE | ID: mdl-20625112

ABSTRACT

BACKGROUND: Apoptosis may contribute to the development of heart failure, but the role of apoptotic signaling initiated by the endoplasmic reticulum in this condition has not been well clarified. METHODS AND RESULTS: In myocardial samples from patients with heart failure, quantitative real-time polymerase chain reaction revealed an increase in messenger RNA for C/EBP homologous protein (CHOP), a transcriptional factor that mediates endoplasmic reticulum-initiated apoptotic cell death. We performed transverse aortic constriction or sham operation on wild-type (WT) and CHOP-deficient mice. The CHOP-deficient mice showed less cardiac hypertrophy, fibrosis, and cardiac dysfunction compared with WT mice at 4 weeks after transverse aortic constriction, although the contractility of isolated cardiomyocytes from CHOP-deficient mice was not significantly different from that in the WT mice. In the hearts of CHOP-deficient mice, phosphorylation of eukaryotic translation initiation factor 2alpha, which may reduce protein translation, was enhanced compared with WT mice. In the hearts of WT mice, CHOP-increased apoptotic cell death with activation of caspase-3 was observed at 4 weeks after transverse aortic constriction. In contrast, CHOP-deficient mice had less apoptotic cell death and lower caspase-3 activation at 4 weeks after transverse aortic constriction. Furthermore, the Bcl2/Bax ratio was decreased in WT mice, whereas this change was significantly blunted in CHOP-deficient mice. Real-time polymerase chain reaction microarray analysis revealed that CHOP could regulate several Bcl2 family members in failing hearts. CONCLUSIONS: We propose the novel concept that CHOP, which may modify protein translation and mediate endoplasmic reticulum-initiated apoptotic cell death, contributes to development of cardiac hypertrophy and failure induced by pressure overload.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Heart Failure/genetics , Animals , Apoptosis/physiology , CCAAT-Enhancer-Binding Proteins/deficiency , Cardiomyopathies/pathology , Cardiomyopathies/surgery , DNA Primers , Humans , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , RNA, Messenger/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction , Stroke Volume
12.
Nat Cell Biol ; 5(9): 781-92, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12907943

ABSTRACT

Excess cellular cholesterol induces apoptosis in macrophages, an event likely to promote progression of atherosclerosis. The cellular mechanism of cholesterol-induced apoptosis is unknown but had previously been thought to involve the plasma membrane. Here we report that the unfolded protein response (UPR) in the endoplasmic reticulum is activated in cholesterol-loaded macrophages, resulting in expression of the cell death effector CHOP. Cholesterol loading depletes endoplasmic reticulum calcium stores, an event known to induce the UPR. Furthermore, endoplasmic reticulum calcium depletion, the UPR, caspase-3 activation and apoptosis are markedly inhibited by selective inhibition of cholesterol trafficking to the endoplasmic reticulum, and Chop-/- macrophages are protected from cholesterol-induced apoptosis. We propose that cholesterol trafficking to endoplasmic reticulum membranes, resulting in activation of the CHOP arm of the UPR, is the key signalling step in cholesterol-induced apoptosis in macrophages.


Subject(s)
Apoptosis/physiology , Cholesterol/toxicity , Coronary Artery Disease/metabolism , Endoplasmic Reticulum/metabolism , Macrophages/metabolism , Protein Folding , Animals , Apoptosis/drug effects , Biological Transport, Active/drug effects , Biological Transport, Active/physiology , CCAAT-Enhancer-Binding Proteins/deficiency , CCAAT-Enhancer-Binding Proteins/genetics , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cells, Cultured , Cholesterol/metabolism , Coronary Artery Disease/physiopathology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/ultrastructure , Female , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Intracellular Membranes/ultrastructure , Macrophages/drug effects , Macrophages/pathology , Mice , Mice, Knockout , Signal Transduction/drug effects , Signal Transduction/physiology , Transcription Factor CHOP , Transcription Factors/deficiency , Transcription Factors/genetics
13.
Aging Cell ; 20(7): e13382, 2021 07.
Article in English | MEDLINE | ID: mdl-34128315

ABSTRACT

Hematopoietic stem cells (HSCs) reside in a quiescent niche to reserve their capacity of self-renewal. Upon hematopoietic injuries, HSCs enter the cell cycle and encounter protein homeostasis problems caused by accumulation of misfolded proteins. However, the mechanism by which protein homeostasis influences HSC function and maintenance remains poorly understood. Here, we show that C/EBP homologous protein (CHOP), demonstrated previously to induces cell death upon unfolded protein response (UPR), plays an important role in HSCs regeneration. CHOP-/- mice showed normal hematopoietic stem and progenitor cell frequencies in steady state. However, when treated with 5-FU, CHOP deficiency resulted in higher survival rates, associated with an increased number of HSCs and reduced level of apoptosis. In serial competitive transplantation experiments, CHOP-/- HSCs showed a dramatic enhancement of repopulation ability and a reduction of protein aggresomes. Mechanistically, CHOP deletion causes reduced ATF3 expression and further leads to decreased protein aggregation and ROS. In addition, CHOP-/- HSCs exhibited an increased resistance to IR-induced DNA damage and improved HSCs homeostasis and function in telomere dysfunctional (G3Terc-/- ) mice. In summary, these findings disclose a new role of CHOP in the regulation of the HSCs function and homeostasis through reducing ATF3 and ROS signaling.


Subject(s)
Activating Transcription Factor 3/metabolism , CCAAT-Enhancer-Binding Proteins/deficiency , Hematopoietic Stem Cells/metabolism , Reactive Oxygen Species/metabolism , Animals , Apoptosis/physiology , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Differentiation , Hematopoietic Stem Cells/cytology , Mice , Protein Deficiency/metabolism
14.
J Exp Med ; 218(3)2021 03 01.
Article in English | MEDLINE | ID: mdl-33616624

ABSTRACT

Frequent outbreaks of viruses have caused a serious threat to public health. Previous evidence has revealed that DNA methylation is correlated with viral infections, but its role in innate immunity remains poorly investigated. Additionally, DNA methylation inhibitors promote IFN-I by upregulating endogenous retrovirus; however, studies of intrinsically demethylated tumors do not support this conclusion. This study found that Uhrf1 deficiency in myeloid cells significantly upregulated Ifnb expression, increasing resistance to viral infection. We performed whole-genome bisulfite sequencing and found that a single-nucleotide methylation site in the Ifnb promoter region disrupted IRF3 recruitment. We used site-specific mutant knock-in mice and a region-specific demethylation tool to confirm that this methylated site plays a critical role in regulating Ifnb expression and antiviral responses. These findings provide essential insight into DNA methylation in the regulation of the innate antiviral immune response.


Subject(s)
Antiviral Agents/metabolism , DNA Methylation/genetics , Immunity, Innate/genetics , Interferon Type I/metabolism , Nucleotides/genetics , Animals , Base Sequence , CCAAT-Enhancer-Binding Proteins/deficiency , CCAAT-Enhancer-Binding Proteins/metabolism , Chromatin/metabolism , CpG Islands/genetics , Cytokines/metabolism , HEK293 Cells , Homeostasis , Humans , Immune System/metabolism , Influenza Vaccines/immunology , Mice , Myeloid Cells/metabolism , Orthomyxoviridae/physiology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Promoter Regions, Genetic/genetics , Signal Transduction , Toll-Like Receptors/agonists , Toll-Like Receptors/metabolism , Transcription, Genetic , Transcriptome/genetics , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/metabolism
15.
J Clin Invest ; 131(13)2021 07 01.
Article in English | MEDLINE | ID: mdl-34032634

ABSTRACT

The role of PI3K and Hippo signaling in chronic pancreatitis (CP) pathogenesis is unclear. Therefore, we assessed the involvement of these pathways in CP by examining the PI3K and Hippo signaling components PTEN and SAV1, respectively. We observed significant decreases in pancreatic PTEN and SAV1 levels in 2 murine CP models: repeated cerulein injection and pancreatic ductal ligation. Additionally, pancreas-specific deletion of Pten and Sav1 (DKO) induced CP in mice. Pancreatic connective tissue growth factor (CTGF) was markedly upregulated in both CP models and DKO mice, and pancreatic CCAAT/enhancer-binding protein-α (CEBPA) expression was downregulated in the CP models. Interestingly, in pancreatic acinar cells (PACs), CEBPA knockdown reduced PTEN and SAV1 and increased CTGF levels in vitro. Furthermore, CEBPA knockdown in PACs induced acinar-to-ductal metaplasia and activation of cocultured macrophages and pancreatic stellate cells. These results were mitigated by CTGF inhibition. CP in DKO mice was also ameliorated by Ctgf gene deletion, and cerulein-induced CP was alleviated by antibody-mediated CTGF neutralization. Finally, we observed significantly decreased PTEN, SAV1, and CEBPA and increased CTGF levels in human CP tissues compared with nonpancreatitis tissues. Taken together, our results indicate that dysregulation of PI3K and Hippo signaling induces CP via CTGF upregulation.


Subject(s)
Connective Tissue Growth Factor/metabolism , Pancreatitis, Chronic/etiology , Pancreatitis, Chronic/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , CCAAT-Enhancer-Binding Proteins/deficiency , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Ceruletide/toxicity , Coculture Techniques , Connective Tissue Growth Factor/antagonists & inhibitors , Connective Tissue Growth Factor/genetics , Disease Models, Animal , Down-Regulation , Hippo Signaling Pathway , Humans , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Pancreatic Stellate Cells/metabolism , Pancreatic Stellate Cells/pathology , Pancreatitis, Chronic/pathology , Signal Transduction , Up-Regulation
16.
Cell Death Dis ; 11(2): 142, 2020 02 21.
Article in English | MEDLINE | ID: mdl-32081844

ABSTRACT

5'-hydroxymethylcytosine (5hmC), an important 5'-cytosine modification, is altered highly in order in male meiotic prophase. However, the regulatory mechanism of this dynamic change and the function of 5hmC in meiosis remain largely unknown. Using a knockout mouse model, we showed that UHRF1 regulated male meiosis. UHRF1 deficiency led to failure of meiosis and male infertility. Mechanistically, the deficiency of UHRF1 altered significantly the meiotic gene profile of spermatocytes. Uhrf1 knockout induced an increase of the global 5hmC level. The enrichment of hyper-5hmC at transcriptional start sites (TSSs) was highly associated with gene downregulation. In addition, the elevated level of the TET1 enzyme might have contributed to the higher 5hmC level in the Uhrf1 knockout spermatocytes. Finally, we reported Uhrf1, a key gene in male meiosis, repressed hyper-5hmC by downregulating TET1. Furthermore, UHRF1 facilitated RNA polymerase II (RNA-pol2) loading to promote gene transcription. Thus our study demonstrated a potential regulatory mechanism of 5hmC dynamic change and its involvement in epigenetic regulation in male meiosis.


Subject(s)
5-Methylcytosine/analogs & derivatives , CCAAT-Enhancer-Binding Proteins/metabolism , Infertility, Male/enzymology , Meiotic Prophase I , Spermatocytes/enzymology , Testis/enzymology , Ubiquitin-Protein Ligases/metabolism , 5-Methylcytosine/metabolism , Animals , CCAAT-Enhancer-Binding Proteins/deficiency , CCAAT-Enhancer-Binding Proteins/genetics , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Down-Regulation , Epigenesis, Genetic , Fertility , Infertility, Male/genetics , Infertility, Male/physiopathology , Male , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA Polymerase II/metabolism , Signal Transduction , Spermatocytes/pathology , Spermatogenesis , Testis/pathology , Testis/physiopathology , Transcriptional Activation , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics
17.
J Exp Med ; 216(12): 2819-2837, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31515281

ABSTRACT

Regulatory T (T reg) cells are required for the maintenance of immune homeostasis. Both TGF-ß signaling and epigenetic modifications are important for Foxp3 induction, but how TGF-ß signaling participates in the epigenetic regulation of Foxp3 remains largely unknown. Here we showed that T cell-specific ablation of Uhrf1 resulted in T reg-biased differentiation in TCR-stimulated naive T cells in the absence of TGF-ß signaling, and these Foxp3+ T cells had a suppressive function. Adoptive transfer of Uhrf1 -/- naive T cells could significantly suppress colitis due to increased iT reg cell generation. Mechanistically, Uhrf1 was induced upon TCR stimulation and participated in the maintenance of DNA methylation patterns of T reg cell-specific genes during cell division, while it was phosphorylated upon TGF-ß stimulation and sequestered outside the nucleus, and ultimately underwent proteasome-dependent degradation. Collectively, our study reveals a novel epigenetic mechanism of TGF-ß-mediated iT reg cell differentiation by modulating Uhrf1 activity and suggests that Uhrf1 may be a potential therapeutic target in inflammatory diseases for generating stable iT reg cells.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , DNA Methylation , Forkhead Transcription Factors/genetics , Signal Transduction , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , CCAAT-Enhancer-Binding Proteins/deficiency , Cell Differentiation , Colitis/etiology , Colitis/metabolism , Colitis/pathology , Epigenesis, Genetic , Gene Expression Profiling , Lymphocyte Activation , Lymphocyte Count , Mice , Mice, Knockout , Phosphorylation , Proteolysis , T-Lymphocytes, Regulatory/immunology , Transcriptome , Ubiquitin-Protein Ligases/deficiency
18.
Neuron ; 36(4): 585-96, 2002 Nov 14.
Article in English | MEDLINE | ID: mdl-12441049

ABSTRACT

The unfolded protein response (UPR) is a eukaryotic signaling pathway linking protein flux through the endoplasmic reticulum to transcription and translational repression. Herein, we demonstrate UPR activation in the leukodystrophy Pelizaeus-Merzbacher disease (PMD) as well as in three mouse models of this disease and transfected fibroblasts expressing mutant protein. The CHOP protein, widely known as a proapoptotic transcription factor, modulates pathogenesis in the mouse models of PMD; however, this protein exhibits antiapoptotic activity. Together, these data show that the UPR has the potential to modulate disease severity in many cells expressing mutant secretory pathway proteins. Thus, PMD represents the first member of a novel class of disparate degenerative diseases for which UPR activation and signaling is the common pathogenic mechanism.


Subject(s)
Eukaryotic Cells/metabolism , Pelizaeus-Merzbacher Disease/metabolism , Protein Folding , Proteins/metabolism , Animals , CCAAT-Enhancer-Binding Proteins/deficiency , CCAAT-Enhancer-Binding Proteins/genetics , Cells, Cultured , Disease Models, Animal , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Eukaryotic Cells/ultrastructure , Humans , Male , Mice , Mice, Knockout , Mutation/genetics , Myelin Basic Protein/metabolism , Myelin Proteolipid Protein/genetics , Myelin Proteolipid Protein/metabolism , Oligodendroglia/metabolism , Oligodendroglia/ultrastructure , Pelizaeus-Merzbacher Disease/genetics , Pelizaeus-Merzbacher Disease/physiopathology , Protein Biosynthesis , Severity of Illness Index , Signal Transduction/genetics , Transcription Factor CHOP , Transcription Factors/deficiency , Transcription Factors/genetics
19.
J Bone Miner Res ; 33(4): 691-703, 2018 04.
Article in English | MEDLINE | ID: mdl-29149533

ABSTRACT

CCAAT/enhancer-binding protein α (C/ebpα) is critical for osteoclastogenesis by regulating osteoclast (OC) lineage commitment and is also important for OC differentiation and function in vitro. However, the role of C/ebpα in postnatal skeletal development has not been reported owing to lethality in C/ebpα-/- mice from hypoglycemia within 8 hours after birth. Herein, we generated conditional knockout mice by deleting the C/ebpα gene in monocyte via LysM-Cre to examine its role in OC differentiation and function. C/ebpαf/f LysM-Cre mice exhibited postnatal osteopetrosis due to impaired osteoclastogenesis, OC lineage priming defects, as well as defective OC differentiation and activity. Furthermore, our ex vivo analysis demonstrated that C/ebpα conditional deletion significantly reduced OC differentiation, maturation, and activity while mildly repressing macrophage development. At the molecular level, C/ebpα deficiency significantly suppresses the expressions of OC genes associated with early stages of osteoclastogenesis as well as genes associated with OC differentiation and activity. We also identified numerous C/ebpα critical cis-regulatory elements on the Cathepsin K promoter that allow C/ebpα to significantly upregulate Cathepsin K expression during OC differentiation and activity. In pathologically induced mouse model of osteoporosis, C/ebpα deficiency can protect mice against ovariectomy-induced bone loss, uncovering a central role for C/ebpα in osteolytic diseases. Collectively, our findings have further established C/ebpα as a promising therapeutic target for bone loss by concurrently targeting OC lineage priming, differentiation, and activity. © 2017 American Society for Bone and Mineral Research.


Subject(s)
CCAAT-Enhancer-Binding Proteins/deficiency , Cell Differentiation , Monocytes/metabolism , Osteoclasts/metabolism , Osteopetrosis/metabolism , Animals , Cathepsin K/biosynthesis , Cathepsin K/genetics , Female , Gene Expression Regulation, Enzymologic , Mice , Mice, Knockout , Monocytes/pathology , Osteoclasts/pathology , Osteolysis/genetics , Osteolysis/metabolism , Osteolysis/pathology , Osteopetrosis/genetics , Osteopetrosis/pathology , Ovariectomy
20.
Cell Rep ; 23(9): 2744-2757, 2018 05 29.
Article in English | MEDLINE | ID: mdl-29847803

ABSTRACT

Transcription factors PU.1 and CEBPA are required for the proper coordination of enhancer activity during granulocytic-monocytic (GM) lineage differentiation to form myeloid cells. However, precisely how these factors control the chronology of enhancer establishment during differentiation is not known. Through integrated analyses of enhancer dynamics, transcription factor binding, and proximal gene expression during successive stages of murine GM-lineage differentiation, we unravel the distinct kinetics by which PU.1 and CEBPA coordinate GM enhancer activity. We find no evidence of a pioneering function of PU.1 during late GM-lineage differentiation. Instead, we delineate a set of enhancers that gain accessibility in a CEBPA-dependent manner, suggesting a pioneering function of CEBPA. Analyses of Cebpa null bone marrow demonstrate that CEBPA controls PU.1 levels and, unexpectedly, that the loss of CEBPA results in an early differentiation block. Taken together, our data provide insights into how PU.1 and CEBPA functionally interact to drive GM-lineage differentiation.


Subject(s)
CCAAT-Enhancer-Binding Proteins/deficiency , Cell Differentiation/genetics , Enhancer Elements, Genetic/genetics , Myeloid Cells/cytology , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Animals , Base Sequence , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Line , Cell Lineage , Chromatin/metabolism , Female , Gene Expression Regulation , Granulocytes/cytology , Granulocytes/metabolism , Mice , Monocytes/cytology , Monocytes/metabolism , Myeloid Cells/metabolism , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL