Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Am J Pathol ; 194(2): 195-208, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37981221

ABSTRACT

miRNAs are small noncoding RNAs that regulate mRNA targets in a cell-specific manner. miR-29 is expressed in murine and human skin, where it may regulate functions in skin repair. Cutaneous wound healing model in miR-29a/b1 gene knockout mice was used to identify miR-29 targets in the wound matrix, where angiogenesis and maturation of provisional granulation tissue was enhanced in response to genetic deletion of miR-29. Consistently, antisense-mediated inhibition of miR-29 promoted angiogenesis in vitro by autocrine and paracrine mechanisms. These processes are likely mediated by miR-29 target mRNAs released upon removal of miR-29 to improve cell-matrix adhesion. One of these, laminin (Lam)-c2 (also known as laminin γ2), was strongly up-regulated during skin repair in the wound matrix of knockout mice. Unexpectedly, Lamc2 was deposited in the basal membrane of endothelial cells in blood vessels forming in the granulation tissue of knockout mice. New blood vessels showed punctate interactions between Lamc2 and integrin α6 (Itga6) along the length of the proto-vessels, suggesting that greater levels of Lamc2 may contribute to the adhesion of endothelial cells, thus assisting angiogenesis within the wound. These findings may be of translational relevance, as LAMC2 was deposited at the leading edge in human wounds, where it formed a basal membrane for endothelial cells and assisted neovascularization. These results suggest a link between LAMC2, improved angiogenesis, and re-epithelialization.


Subject(s)
Laminin , MicroRNAs , Humans , Animals , Mice , Laminin/genetics , Endothelial Cells , Signal Transduction/physiology , MicroRNAs/genetics , Skin , Mice, Knockout
2.
Cardiovasc Res ; 119(9): 1869-1882, 2023 08 07.
Article in English | MEDLINE | ID: mdl-36804807

ABSTRACT

AIMS: Endothelial erosion of plaques is responsible for ∼30% of acute coronary syndromes (ACS). Smoking is a risk factor for plaque erosion, which most frequently occurs on the upstream surface of plaques where the endothelium experiences elevated shear stress. We sought to recreate these conditions in vitro to identify potential pathological mechanisms that might be of relevance to plaque erosion. METHODS AND RESULTS: Culturing human coronary artery endothelial cells (HCAECs) under elevated flow (shear stress of 7.5 Pa) and chronically exposing them to cigarette smoke extract (CSE) and tumour necrosis factor-alpha (TNFα) recapitulated a defect in HCAEC adhesion, which corresponded with augmented Nrf2-regulated gene expression. Pharmacological activation or adenoviral overexpression of Nrf2 triggered endothelial detachment, identifying Nrf2 as a mediator of endothelial detachment. Growth/Differentiation Factor-15 (GDF15) expression was elevated in this model, with protein expression elevated in the plasma of patients experiencing plaque erosion compared with plaque rupture. The expression of two Nrf2-regulated genes, OSGIN1 and OSGIN2, was increased by CSE and TNFα under elevated flow and was also elevated in the aortas of mice exposed to cigarette smoke in vivo. Knockdown of OSGIN1&2 inhibited Nrf2-induced cell detachment. Overexpression of OSGIN1&2 induced endothelial detachment and resulted in cell cycle arrest, induction of senescence, loss of focal adhesions and actin stress fibres, and disturbed proteostasis mediated in part by HSP70, restoration of which reduced HCAEC detachment. In ACS patients who smoked, blood concentrations of HSP70 were elevated in plaque erosion compared with plaque rupture. CONCLUSION: We identified a novel Nrf2-OSGIN1&2-HSP70 axis that regulates endothelial adhesion, elevated GDF15 and HSP70 as biomarkers for plaque erosion in patients who smoke, and two therapeutic targets that offer the potential for reducing the risk of plaque erosion.


Subject(s)
Cigarette Smoking , Plaque, Atherosclerotic , Humans , Animals , Mice , Tumor Necrosis Factor-alpha/pharmacology , Endothelial Cells/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Nicotiana/metabolism , Endothelium/metabolism
3.
Nucleic Acids Res ; 49(7): 3748-3763, 2021 04 19.
Article in English | MEDLINE | ID: mdl-33764436

ABSTRACT

Epigenetic regulation of cell and tissue function requires the coordinated action of transcription factors. However, their combinatorial activities during regeneration remain largely unexplored. Here, we discover an unexpected interaction between the cytoprotective transcription factor NRF2 and p63- a key player in epithelial morphogenesis. Chromatin immunoprecipitation combined with sequencing and reporter assays identifies enhancers and promoters that are simultaneously activated by NRF2 and p63 in human keratinocytes. Modeling of p63 and NRF2 binding to nucleosomal DNA suggests their chromatin-assisted interaction. Pharmacological and genetic activation of NRF2 increases NRF2-p63 binding to enhancers and promotes keratinocyte proliferation, which involves the common NRF2-p63 target cyclin-dependent kinase 12. These results unravel a collaborative function of NRF2 and p63 in the control of epidermal renewal and suggest their combined activation as a strategy to promote repair of human skin and other stratified epithelia.


Subject(s)
Keratinocytes , NF-E2-Related Factor 2/physiology , Skin , Transcription Factors/physiology , Tumor Suppressor Proteins/physiology , Animals , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinases/metabolism , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Mice , Skin/cytology , Skin/metabolism
4.
Biochem Soc Trans ; 48(6): 2839-2850, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33170265

ABSTRACT

The search for epidermal stem cells has gained the momentum as they possess unique biological characteristics and a potential in regeneration therapies. Several transcription factors and miRNAs have been identified as epidermal stem cell markers. However, the separation of epidermal stem cells from their progeny remains challenging. The introduction of single-cell transcriptomics pointed to the high degree of heterogeneity in epidermal stem cells imbedded within subpopulations of keratinocytes. Pseudotime inference, RNA velocity, and cellular entropy further enhanced our knowledge of stem cells, allowing for the discovery of the epidermal stem cell plasticity. We explore the main findings that lead to the discovery of the plastic trait within the epidermal stem cells and the implications of cell plasticity in regenerative medicine.


Subject(s)
Epidermal Cells/cytology , Gene Expression Regulation , Regenerative Medicine/methods , Skin Physiological Phenomena , Skin/metabolism , Stem Cells/cytology , Transcriptome , Animals , Cell Differentiation , Cell Proliferation , Epidermis/metabolism , Humans , Integrins/metabolism , Keratinocytes/cytology , Mice , RNA/metabolism , Regeneration , Signal Transduction , Time Factors , Transcription Factors/metabolism
5.
Biotechniques ; 65(2): 97-100, 2018 08.
Article in English | MEDLINE | ID: mdl-30091388

ABSTRACT

RNA synthesis can be detected by 5-ethynyl uridine (EU) incorporation and click chemistry. Despite identifying a fundamental functional process, this technique has yet to be widely applied to complex human tissue systems. By incorporating EU into human hair follicle (HF) organs cultured ex vivo, nascent RNA synthesis was detected in situ. EU differentially incorporated across the HF epithelium. Interestingly, RNA synthesis did not correlate with protein synthesis, proliferation or epithelial progenitor cell marker expression. By treating human HFs with the cytotoxic cell cycle inhibitor (R)-CR8, which inhibits transcriptional regulators CDK7 and CDK9, it was further shown that this technique can be used to sensitively detect changes in global RNA synthesis in situ. Together, this work delineates new insights into nascent RNA synthesis within a human (mini)- organ and describes a novel read-out parameter that will enrich future ex vivo human tissue research studies.


Subject(s)
Click Chemistry , Hair Follicle/metabolism , Microscopy, Fluorescence , Organ Culture Techniques , RNA/analysis , Uridine/analogs & derivatives , Cell Proliferation , Click Chemistry/methods , Gene Expression Regulation , Hair Follicle/cytology , Humans , Microscopy, Fluorescence/methods , Organ Culture Techniques/methods , RNA/genetics , Transcription, Genetic , Uridine/genetics
6.
J Immunol ; 196(11): 4663-70, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27183581

ABSTRACT

The Nrf2 transcription factor is well known for its cytoprotective functions through regulation of genes involved in the detoxification of reactive oxygen species or toxic compounds. Therefore, activation of Nrf2 is a promising strategy for the protection of tissues from various types of insults and for cancer prevention. However, recent studies revealed a proinflammatory activity of activated Nrf2 and a stimulating effect on epithelial cell proliferation, but the underlying mechanisms of action and the responsible target genes are largely unknown. Using a combination of gene expression profiling, chromatin immunoprecipitation, and targeted proteomics via selected reaction monitoring, we show that the gene encoding the proinflammatory cytokine IL-36γ is a novel direct target of Nrf2 in keratinocytes and hepatocytes in vitro and in vivo. As a consequence, upregulation of IL-36γ expression occurred upon genetic or pharmacological activation of Nrf2 in the epidermis and in the normal and regenerating liver. Functional in vitro studies demonstrate that IL-36γ directly stimulates proliferation of keratinocytes. In particular, it induces expression of keratinocyte mitogens in fibroblasts, suggesting that the Nrf2-IL-36γ axis promotes keratinocyte proliferation through a double paracrine loop. These results provide mechanistic insight into Nrf2 action in the control of inflammation and cell proliferation through regulation of a proinflammatory cytokine with a key function in various inflammatory diseases.


Subject(s)
Autocrine Communication , Cell Proliferation , Interleukin-1/metabolism , Keratinocytes/cytology , Keratinocytes/metabolism , NF-E2-Related Factor 2/metabolism , Paracrine Communication , Animals , Cells, Cultured , Interleukin-1/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Transgenic
7.
Mol Neurobiol ; 53(6): 4210-4225, 2016 08.
Article in English | MEDLINE | ID: mdl-26215835

ABSTRACT

The transforming growth factor-ß (TGF-ß) family member activin A exerts multiple neurotrophic and protective effects in the brain. Activin also modulates cognitive functions and affective behavior and is a presumed target of antidepressant therapy. Despite its important role in the injured and intact brain, the mechanisms underlying activin effects in the CNS are still largely unknown. Our goal was to identify the first target genes of activin signaling in the hippocampus in vivo. Electroconvulsive seizures, a rodent model of electroconvulsive therapy in humans, were applied to C57BL/6J mice to elicit a strong increase in activin A signaling. Chromatin immunoprecipitation experiments with hippocampal lysates subsequently revealed that binding of SMAD2/3, the intracellular effectors of activin signaling, was significantly enriched at the Pmepa1 gene, which encodes a negative feedback regulator of TGF-ß signaling in cancer cells, and at the Kdm6b gene, which encodes an epigenetic regulator promoting transcriptional plasticity. Underlining the significance of these findings, activin treatment also induced PMEPA1 and KDM6B expression in human forebrain neurons generated from embryonic stem cells suggesting interspecies conservation of activin effects in mammalian neurons. Importantly, physiological stimuli such as provided by environmental enrichment proved already sufficient to engender a rapid and significant induction of activin signaling concomitant with an upregulation of Pmepa1 and Kdm6b expression. Taken together, our study identified the first target genes of activin signaling in the brain. With the induction of Kdm6b expression, activin is likely to gain impact on a presumed epigenetic regulator of activity-dependent neuronal plasticity.


Subject(s)
Activins/metabolism , Behavior, Animal , Electrophysiological Phenomena , Jumonji Domain-Containing Histone Demethylases/metabolism , Membrane Proteins/metabolism , Signal Transduction , Animals , Base Sequence , Cell Line , Electroshock , Female , Hippocampus/metabolism , Humans , Male , Mice, Inbred C57BL , Neurons/metabolism , Smad Proteins/metabolism , Up-Regulation
8.
Biochem Soc Trans ; 43(4): 595-601, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26551699

ABSTRACT

The nuclear factor erythroid 2-related factor 2 (NRF2) transcription factor is a key player in the cellular antioxidant response and it also controls various other functions in a cell-type specific manner. Due to these key functions, a tight control of NRF2 expression and activity is essential. This regulation is exerted at multiple levels, including transcriptional regulation and proteasomal degradation. Recent studies revealed important roles of miRNAs (miRs) in the control of NRF2 activity through direct targeting of the NRF2 mRNA and of mRNAs encoding proteins that control the level and activity of NRF2. In addition, NRF2 itself has been identified as a regulator of miRs, which exert some of the functions of NRF2 in metabolic regulation and also novel functions in the regulation of cell adhesion. Here, we summarize the roles and mechanisms of action of miRs in the regulation of NRF2 activity and as downstream effectors of this transcription factor.


Subject(s)
MicroRNAs/genetics , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Gene Expression Regulation , Humans , Kidney/metabolism , Liver/metabolism , Proteasome Endopeptidase Complex/metabolism
9.
Nat Commun ; 5: 5099, 2014 Oct 06.
Article in English | MEDLINE | ID: mdl-25283360

ABSTRACT

The Nrf2 transcription factor controls the expression of genes involved in the antioxidant defense system. Here, we identified Nrf2 as a novel regulator of desmosomes in the epidermis through the regulation of microRNAs. On Nrf2 activation, expression of miR-29a and miR-29b increases in cultured human keratinocytes and in mouse epidermis. Chromatin immunoprecipitation identified the Mir29ab1 and Mir29b2c genes as direct Nrf2 targets in keratinocytes. While binding of Nrf2 to the Mir29ab1 gene activates expression of miR-29a and -b, the Mir29b2c gene is silenced by DNA methylation. We identified desmocollin-2 (Dsc2) as a major target of Nrf2-induced miR-29s. This is functionally important, since Nrf2 activation in keratinocytes of transgenic mice causes structural alterations of epidermal desmosomes. Furthermore, the overexpression of miR-29a/b or knockdown of Dsc2 impairs the formation of hyper-adhesive desmosomes in keratinocytes, whereas Dsc2 overexpression has the opposite effect. These results demonstrate that a novel Nrf2-miR-29-Dsc2 axis controls desmosome function and cutaneous homeostasis.


Subject(s)
Desmocollins/metabolism , Desmosomes/metabolism , Keratinocytes/cytology , MicroRNAs/metabolism , NF-E2-Related Factor 2/metabolism , Animals , Base Sequence , Cell Line, Tumor , Chromatin Immunoprecipitation , CpG Islands , DNA Methylation , Gene Expression Regulation , Gene Silencing , Homeostasis , Humans , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic , Molecular Sequence Data , Skin/metabolism
10.
EMBO Mol Med ; 6(4): 442-57, 2014 04.
Article in English | MEDLINE | ID: mdl-24503019

ABSTRACT

The transcription factor Nrf2 is a key regulator of the cellular stress response, and pharmacological Nrf2 activation is a promising strategy for skin protection and cancer prevention. We show here that prolonged Nrf2 activation in keratinocytes causes sebaceous gland enlargement and seborrhea in mice due to upregulation of the growth factor epigen, which we identified as a novel Nrf2 target. This was accompanied by thickening and hyperkeratosis of hair follicle infundibula. These abnormalities caused dilatation of infundibula, hair loss, and cyst development upon aging. Upregulation of epigen, secretory leukocyte peptidase inhibitor (Slpi), and small proline-rich protein 2d (Sprr2d) in hair follicles was identified as the likely cause of infundibular acanthosis, hyperkeratosis, and cyst formation. These alterations were highly reminiscent to the phenotype of chloracne/"metabolizing acquired dioxin-induced skin hamartomas" (MADISH) patients. Indeed, SLPI, SPRR2, and epigen were strongly expressed in cysts of MADISH patients and upregulated by dioxin in human keratinocytes in an NRF2-dependent manner. These results identify novel Nrf2 activities in the pilosebaceous unit and point to a role of NRF2 in MADISH pathogenesis.


Subject(s)
Chloracne/metabolism , Keratinocytes/metabolism , NF-E2-Related Factor 2/metabolism , Animals , Cells, Cultured , Chloracne/genetics , Disease Models, Animal , Epigen/genetics , Epigen/metabolism , Hair Follicle/metabolism , Humans , Mice , Mice, Transgenic , NF-E2-Related Factor 2/genetics , Secretory Leukocyte Peptidase Inhibitor/genetics , Secretory Leukocyte Peptidase Inhibitor/metabolism
11.
Hepatology ; 60(2): 670-8, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24310875

ABSTRACT

UNLABELLED: The nuclear factor erythroid-derived 2, like 2 (Nrf2) transcription factor is a key regulator of the antioxidant defense system, and pharmacological activation of Nrf2 is a promising strategy for prevention of toxin-induced liver damage. However, the consequences of Nrf2 activation on liver regeneration (LR) have not been determined. To address this question, we generated mice expressing a constitutively active Nrf2 (caNrf2) mutant in hepatocytes. Expression of the transgene did not affect liver homeostasis. Surprisingly, however, there was no beneficial effect of Nrf2 activation on CCl4 -induced liver injury and fibrosis. Most important, LR after partial hepatectomy was impaired in caNrf2-transgenic mice as a result of delayed hepatocyte proliferation and enhanced apoptosis of these cells after liver injury. Mechanistically, this involved up-regulation of the cyclin-dependent kinase inhibitor p15 and the proapoptotic protein Bcl2l11 (Bim). Using chromatin immunoprecipitation, we show that the p15 and Bcl2l11 genes are direct targets of Nrf2, which are activated under hyperproliferative conditions in the liver. CONCLUSION: Activated Nrf2 delays proliferation and induces apoptosis of hepatocytes in the regenerating liver. These negative effects of Nrf2 activation on LR should be considered when Nrf2-activating compounds are used for prevention of liver damage.


Subject(s)
Apoptosis/physiology , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/metabolism , Liver Regeneration/physiology , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Carbon Tetrachloride Poisoning/genetics , Carbon Tetrachloride Poisoning/metabolism , Cell Cycle/physiology , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p15/metabolism , Disease Models, Animal , Hepatocytes/physiology , Homeostasis/physiology , Insulin-Like Growth Factor I/metabolism , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Proto-Oncogene Proteins/metabolism , Receptor, Notch1/metabolism
12.
Hepatology ; 57(5): 2004-13, 2013 May.
Article in English | MEDLINE | ID: mdl-23300120

ABSTRACT

UNLABELLED: Functions of p53 during mitosis reportedly include prevention of polyploidy and transmission of aberrant chromosomes. However, whether p53 plays these roles during genomic surveillance in vivo and, if so, whether this is done via direct or indirect means remain unknown. The ability of normal, mature hepatocytes to respond to stimuli, reenter the cell cycle, and regenerate liver mass offers an ideal setting to assess mitosis in vivo. In quiescent liver, normally high ploidy levels in adult mice increased with loss of p53. Following partial hepatectomy, p53(-/-) hepatocytes exhibited early entry into the cell cycle and prolonged proliferation with an increased number of polyploid mitoses. Ploidy levels increased during regeneration of both wild-type (WT) and p53(-/-) hepatocytes, but only WT hepatocytes were able to dynamically resolve ploidy levels and return to normal by the end of regeneration. We identified multiple cell cycle and mitotic regulators, including Foxm1, Aurka, Lats2, Plk2, and Plk4, as directly regulated by chromatin interactions of p53 in vivo. Over a time course of regeneration, direct and indirect regulation of expression by p53 is mediated in a gene-specific manner. CONCLUSION: Our results show that p53 plays a role in mitotic fidelity and ploidy resolution in hepatocytes of normal and regenerative liver.


Subject(s)
Liver/pathology , Mitosis/physiology , Ploidies , Transcription, Genetic/physiology , Tumor Suppressor Protein p53/physiology , Animals , Cell Cycle/physiology , Cell Proliferation , Hepatectomy , Liver/physiology , Liver/surgery , Liver Regeneration/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
13.
PLoS Genet ; 8(6): e1002770, 2012.
Article in English | MEDLINE | ID: mdl-22737085

ABSTRACT

Gene duplication is a powerful driver of evolution. Newly duplicated genes acquire new roles that are relevant to fitness, or they will be lost over time. A potential path to functional relevance is mutation of the coding sequence leading to the acquisition of novel biochemical properties, as analyzed here for the highly homologous paralogs Foxa1 and Foxa2 transcriptional regulators. We determine by genome-wide location analysis (ChIP-Seq) that, although Foxa1 and Foxa2 share a large fraction of binding sites in the liver, each protein also occupies distinct regulatory elements in vivo. Foxa1-only sites are enriched for p53 binding sites and are frequently found near genes important to cell cycle regulation, while Foxa2-restricted sites show only a limited match to the forkhead consensus and are found in genes involved in steroid and lipid metabolism. Thus, Foxa1 and Foxa2, while redundant during development, have evolved divergent roles in the adult liver, ensuring the maintenance of both genes during evolution.


Subject(s)
DNA-Binding Proteins , Hepatocyte Nuclear Factor 3-alpha , Hepatocyte Nuclear Factor 3-beta , Liver , Transcription, Genetic , Amino Acid Sequence , Animals , Binding Sites , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryonic Development/genetics , Gene Duplication , Gene Expression Regulation , Genes, p53/genetics , Genome , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-alpha/metabolism , Hepatocyte Nuclear Factor 3-beta/genetics , Hepatocyte Nuclear Factor 3-beta/metabolism , Liver/growth & development , Liver/metabolism , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation , Nucleotide Motifs , Sequence Homology
14.
Toxicol Appl Pharmacol ; 255(1): 76-85, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21672546

ABSTRACT

2'-3-dimethyl-4-aminoazobenzene (ortho-aminoazotoluene, OAT) is an azo dye and a rodent carcinogen that has been evaluated by the International Agency for Research on Cancer (IARC) as a possible (class 2B) human carcinogen. Its mechanism of action remains unclear. We examined the role of the xenobiotic receptor Constitutive Androstane Receptor (CAR, NR1I3) as a mediator of the effects of OAT. We found that OAT increases mouse CAR (mCAR) transactivation in a dose-dependent manner. This effect is specific because another closely related azo dye, 3'-methyl-4-dimethyl-aminoazobenzene (3'MeDAB), did not activate mCAR. Real-time Q-PCR analysis in wild-type C57BL/6 mice revealed that OAT induces the hepatic mRNA expression of the following CAR target genes: Cyp2b10, Cyp2c29, Cyp3a11, Ugt1a1, Mrp4, Mrp2 and c-Myc. CAR-null (Car(-/-)) mice showed no increased expression of these genes following OAT treatment, demonstrating that CAR is required for their OAT dependent induction. The OAT-induced CAR-dependent increase of Cyp2b10 and c-Myc expression was confirmed by Western blotting. Immunohistochemistry analysis of wild-type and Car(-/-) livers showed that OAT did not acutely induce hepatocyte proliferation, but at much later time points showed an unexpected CAR-dependent proliferative response. These studies demonstrate that mCAR is an OAT xenosensor, and indicate that at least some of the biological effects of this compound are mediated by this nuclear receptor.


Subject(s)
Receptors, Cytoplasmic and Nuclear/drug effects , o-Aminoazotoluene/toxicity , Animals , Aryl Hydrocarbon Hydroxylases/genetics , Cell Proliferation/drug effects , Constitutive Androstane Receptor , Cytochrome P450 Family 2 , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/physiology , Humans , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-myc/genetics , RNA, Messenger/analysis , Receptors, Cytoplasmic and Nuclear/physiology , Steroid Hydroxylases/genetics
15.
Int J Biochem Cell Biol ; 43(2): 189-97, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20307684

ABSTRACT

An increasing demand for new strategies in cancer prevention and regenerative medicine requires a better understanding of molecular mechanisms that control cell proliferation in tissue-specific manner. Regenerating liver is a unique model allowing use of biochemical, genetic, and engineering tools to uncover molecular mechanisms and improve treatment of hepatic cancers, liver failure, and fibrotic disease. Molecular mechanisms of liver regeneration involve extra- and intracellular factors to activate transcription of genes normally silenced in quiescent liver. While many upstream signaling pathways of the regenerating liver have been extensively studied, our knowledge of the downstream effectors, transcription factors (TFs), remains limited. This review describes consecutive engagement of pre-existing and de novo synthesized TFs, as cascades that regulate expression of growth-related and metabolic genes during liver regeneration after partial hepatectomy in mice. Several previously recognized regulators of regenerating liver are described in the light of recently identified co-activator and co-repressor complexes that interact with primary DNA-binding TFs. Published results of gene expression and chromatin immunoprecipitation analyses, as well as studies of transgenic mouse models, are used to emphasize new potential regulators of transcription during liver regeneration. Finally, a more detailed description of newly identified transcriptional regulators of liver regeneration illustrates the tightly regulated balance of proliferative and metabolic responses to partial hepatectomy.


Subject(s)
Gene Expression Regulation , Liver Regeneration , Animals , Cell Communication , Cell Cycle , Cell Proliferation , Hepatectomy , Hepatocytes/physiology , Humans , Mice , Receptors, Cytoplasmic and Nuclear/physiology , Signal Transduction
16.
Prog Mol Biol Transl Sci ; 97: 201-27, 2010.
Article in English | MEDLINE | ID: mdl-21074734

ABSTRACT

The remarkable capacity of the liver to regenerate after severe injury or disease has excited interest for centuries. The goal of harnessing this process in treatment of liver disease, and the appreciation of the parallels between regeneration and tumor development in the liver, remain a major driver for research in this area. Studies of liver regeneration as a model system offer a view of intricate and precisely timed regulatory pathways that drive the process toward completion. Successful regeneration of the liver mass demands a hierarchal and well-controlled balance between proliferative and metabolic functions, which is orchestrated by signaling and regulation of transcription factors. Control and regulation of these cascades of transcriptional activities, necessary for induction, renewal, and cessation of liver growth, are the focus of this chapter.


Subject(s)
Gene Expression Regulation , Liver Regeneration/genetics , Transcription, Genetic , Animals , Cell Proliferation , Humans , Models, Biological , Transcription Factors/metabolism
17.
Hepatology ; 52(3): 1023-32, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20564353

ABSTRACT

UNLABELLED: The p53 family of proteins regulates the expression of target genes that promote cell cycle arrest and apoptosis, which may be linked to cellular growth control as well as tumor suppression. Within the p53 family, p53 and the transactivating p73 isoform (TA-p73) have hepatic-specific functions in development and tumor suppression. Here, we determined TA-p73 interactions with chromatin in the adult mouse liver and found forkhead box O3 (Foxo3) to be one of 158 gene targets. Global profiling of hepatic gene expression in the regenerating liver versus the quiescent liver revealed specific, functional categories of genes regulated over the time of regeneration. Foxo3 is the most responsive gene among transcription factors with altered expression during regenerative cellular proliferation. p53 and TA-p73 bind a Foxo3 p53 response element (p53RE) and maintain active expression in the quiescent liver. During regeneration of the liver, the binding of p53 and TA-p73, the recruitment of acetyltransferase p300, and the active chromatin structure of Foxo3 are disrupted along with a loss of Foxo3 expression. In agreement with the loss of Foxo3 transcriptional activation, a decrease in histone activation marks (dimethylated histone H3 at lysine 4, acetylated histone H3 at lysine 14, and acetylated H4) at the Foxo3 p53RE was detected after partial hepatectomy in mice. These parameters of Foxo3 regulation are reestablished with the completion of liver growth and regeneration and support a temporary suspension of p53 and TA-p73 regulatory functions in normal cells during tissue regeneration. p53-dependent and TA-p73-dependent activation of Foxo3 was also observed in mouse embryonic fibroblasts and in mouse hepatoma cells overexpressing p53, TA-p73alpha, and TA-p73beta isoforms. CONCLUSION: p53 and p73 directly bind and activate the expression of the Foxo3 gene in the adult mouse liver and murine cell lines. p53, TA-p73, and p300 binding and Foxo3 expression decrease during liver regeneration, and this suggests a critical growth control mechanism mediated by these transcription factors in vivo.


Subject(s)
Forkhead Transcription Factors/metabolism , Liver Regeneration/physiology , Liver/metabolism , Nuclear Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cells, Cultured , Fibroblasts/metabolism , Fibroblasts/pathology , Forkhead Box Protein O3 , Hepatectomy , Histones/metabolism , Liver/pathology , Liver/surgery , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, Inbred C57BL , Models, Animal , p300-CBP Transcription Factors/metabolism
18.
Cell Cycle ; 7(21): 3362-70, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18948750

ABSTRACT

Ceramide is a sphingolipid that activates stress kinases such as p38 and c-JUN N-Terminal Kinase (JNK). Though Chronic Myelogenous Leukemia (CML) derived K562 cells resist killing by short chain C2-ceramide, we report here that longer chain C6-ceramide promotes apoptosis in these cells. C6-ceramide induces cleavage of Caspase-8 and Caspase-9, but only Caspase-8 is required for apoptosis. The sphingolipid killed CML derived KBM5 cells and, to a lesser extent, imatinib-resistant KBM5-STI cells suggesting that BCR-ABL can not completely block C6-ceramide-induced apoptosis but the kinase may regulate the process. BCR-ABL is known to suppress Protein Phosphatase 2A (PP2A) in CML cells. While C6-ceramide can activate PP2A in acute leukemia cells, the sphingolipid did not activate the phosphatase in K562 cells. C6-ceramide did not activate p38 kinase but did promote JNK activation and phosphorylation of JUN. Inhibition of JNK by pharmacological agent protected K562 cells from C6-ceramide suggesting that JNK plays an essential role in C6-ceramide mediated apoptosis. Furthermore, the sphingolipid promoted MCL-1 phosphorylation by a mechanism that, at least in part, involves JNK. The findings presented here suggest that Caspase-8, JNK, and perhaps MCL-1 may play important roles in regulating cell death and may represent new targets for therapeutic strategies for CML.


Subject(s)
Apoptosis/drug effects , Caspase 8/metabolism , Ceramides/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Benzamides , Drug Resistance, Neoplasm/drug effects , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Fusion Proteins, bcr-abl/metabolism , Humans , Imatinib Mesylate , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , K562 Cells , Myeloid Cell Leukemia Sequence 1 Protein , Phosphorylation/drug effects , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyrimidines/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
19.
J Biol Chem ; 283(51): 35474-85, 2008 Dec 19.
Article in English | MEDLINE | ID: mdl-18957415

ABSTRACT

Protein phosphatase 2A (PP2A) is a heterotrimer comprising catalytic, scaffold, and regulatory (B) subunits. There are at least 21 B subunit family members. Thus PP2A is actually a family of enzymes defined by which B subunit is used. The B56 family member B56alpha is a phosphoprotein that regulates dephosphorylation of BCL2. The stress kinase PKR has been shown to phosphorylate B56alpha at serine 28 in vitro, but it has been unclear how PKR might regulate the BCL2 phosphatase. In the present study, PKR regulation of B56alpha in REH cells was examined, because these cells exhibit robust BCL2 phosphatase activity. PKR was found to be basally active in REH cells as would be predicted if the kinase supports B56alpha-mediated dephosphorylation of BCL2. Suppression of PKR promoted BCL2 phosphorylation with concomitant loss of B56alpha phosphorylation at serine 28 and inhibition of mitochondrial PP2A activity. PKR supports stress signaling in REH cells, as suppression of PKR promoted chemoresistance to etoposide. Suppression of PKR promoted B56alpha proteolysis, which could be blocked by a proteasome inhibitor. However, the mechanism by which PKR supports B56alpha protein does not involve PKR-mediated phosphorylation of the B subunit at serine 28 but may involve eIF2alpha activation of AKT. Phosphorylation of serine 28 by PKR promotes mitochondrial localization of B56alpha, because wild-type but not mutant S28A B56alpha promoted mitochondrial PP2A activity. Cells expressing wild-type B56alpha but not S28A B56alpha were sensitized to etoposide. These results suggest that PKR regulates B56alpha-mediated PP2A signaling in REH cells.


Subject(s)
Eukaryotic Initiation Factor-2B/metabolism , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction/physiology , eIF-2 Kinase/metabolism , Amino Acid Substitution , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Etoposide/pharmacology , Eukaryotic Initiation Factor-2B/genetics , Humans , Mutation, Missense , Phosphorylation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Protein Phosphatase 2/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , eIF-2 Kinase/genetics
20.
Mol Cancer Ther ; 7(5): 1130-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18483301

ABSTRACT

The initial success of the first synthetic bcr-abl kinase inhibitor imatinib has been dampened by the emergence of imatinib-resistant disease in blast crisis chronic myeloid leukemia. Here, we report that the novel triterpenoid methyl-2-cyano-3,12-dioxooleana-1,9-diene-28-oate (CDDO-Me) potently induced cytotoxicity in imatinib-resistant KBM5 cells expressing the T315I mutation of bcr-abl (24-h EC50, 540 nmol/L). In long-term culture, CDDO-Me abrogated the growth of human parental KBM5 and KBM5-STI cells with 96-h IC50 of 205 and 221 nmol/L, respectively. In addition, CDDO-Me rapidly decreased the viability of murine lymphoid Ba/F3 cells expressing wild-type p210 as well as the imatinib-resistant E255K and T315I mutations of bcr-abl. The low-dose effects of CDDO-Me are associated with inhibition of mitochondrial oxygen consumption, whereas the cytotoxic effects appear to be mediated by a rapid and selective depletion of mitochondrial glutathione that accompanies the increased generation of reactive oxygen species and mitochondrial dysfunction. Interestingly, the mitochondriotoxic effects of CDDO-Me are followed by the rapid autophagocytosis of intracellular organelles or the externalization of phosphatidylserine in different cell types. We conclude that alterations in mitochondrial function by CDDO-Me can result in autophagy or apoptosis of chronic myeloid leukemia cells regardless of the mutational status of bcr-abl. CDDO-Me is in clinical trials and shows signs of clinical activity, with minimal side effects and complete lack of cardiotoxicity. Studies in leukemias are in preparation.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Autophagy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Mitochondria/drug effects , Oleanolic Acid/analogs & derivatives , Benzamides , Dose-Response Relationship, Drug , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mitochondria/metabolism , Oleanolic Acid/pharmacology , Oxidation-Reduction , Oxygen/metabolism , Piperazines/pharmacology , Pyrimidines/pharmacology , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...