Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Mol Neurobiol ; 57(3): 1446-1458, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31758402

ABSTRACT

Vascular mechanisms underlying the adverse effects that depression and stress-related mental disorders have on stroke outcome are only partially understood. Identifying the transcriptomic signature of chronic stress in endothelium harvested from the ischemic brain is an important step towards elucidating the biological processes involved. Here, we subjected male 129S6/SvEv mice to a 28-day model of chronic stress. The ischemic lesion was quantified after 30 min filamentous middle cerebral artery occlusion (MCAo) and 48 h reperfusion by T2-weighted MRI. RNA sequencing was used to profile transcriptomic changes in cerebrovascular endothelial cells (ECs) from the infarct. Mice subjected to the stress procedure displayed reduced weight gain, increased adrenal gland weight, and increased hypothalamic FKBP5 mRNA and protein expression. Chronic stress conferred increased lesion volume upon MCAo. Stress-exposed mice showed a higher number of differentially expressed genes between ECs isolated from the ipsilateral and contralateral hemisphere than control mice. The genes in question are enriched for roles in biological processes closely linked to endothelial proliferation and neoangiogenesis. MicroRNA-34a was associated with nine of the top 10 biological process Gene Ontology terms selectively enriched in ECs from stressed mice. Moreover, expression of mature miR-34a-5p and miR-34a-3p in ischemic brain tissue was positively related to infarct size and negatively related to sirtuin 1 (Sirt1) mRNA transcription. In conclusion, this study represents the first EC-specific transcriptomic analysis of chronic stress in brain ischemia. The stress signature uncovered relates to worse stroke outcome and is directly relevant to endothelial mechanisms in the pathogenesis of stroke.


Subject(s)
Brain Ischemia/metabolism , Brain/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , Ischemia/metabolism , Animals , Brain Ischemia/pathology , Disease Models, Animal , Endothelium, Vascular/metabolism , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Magnetic Resonance Imaging/methods , Male , Mice
2.
Neuroscience ; 416: 20-29, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31356897

ABSTRACT

In the adult hippocampal dentate gyrus (DG), the majority of newly generated cells are eliminated by apoptotic mechanisms. The apoptosis repressor with caspase recruitment domain (ARC), encoded by the Nol3 gene, is a potent and multifunctional death repressor that inhibits both death receptor and mitochondrial apoptotic signaling. The aim of the present study was to parse the role of ARC in the development of new granule cell neurons. Nol3 gene expression as revealed by in situ hybridization is present in the entire dentate granule cell layer. Moreover, a comparison of Nol3 expression between FACS-sorted Sox2-positive neural stem cells and Doublecortin (DCX)-positive immature neurons demonstrates upregulation of Nol3 during neurogenesis. Using ARC-deficient mice, we show that proliferation and survival of BrdU birth-dated cells are strongly reduced in the absence of ARC while neuronal-glial fate choice is not affected. Both the number of DCX-positive cells and the number of calretinin (CR)-positive immature postmitotic neurons are reduced in the hippocampus of ARC-/- mice. ARC knockout is not associated with increased numbers of microglia or with microglia activation. However, hippocampal brain-derived neurotrophic factor (BDNF) protein content is significantly increased in ARC-/- mice, possibly representing a compensatory response. Collectively, our results suggest that ARC plays a critical cell-autonomous role in preventing cell death during adult granule cell neogenesis.


Subject(s)
Apoptosis/physiology , Caspase Activation and Recruitment Domain/physiology , Neurogenesis/physiology , Neurons/metabolism , AIDS-Related Complex/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Proliferation/physiology , Doublecortin Protein , Hippocampus/metabolism , Mice, Knockout , Neural Stem Cells/metabolism , Neuroglia/metabolism
3.
J Mol Med (Berl) ; 97(8): 1127-1138, 2019 08.
Article in English | MEDLINE | ID: mdl-31147725

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) control the expression of genes involved in glucose homeostasis, lipid metabolism, inflammation, and cell differentiation. Here, we analyzed the effects of aleglitazar, a dual PPARα and PPARγ agonist with balanced affinity for either subtype, on subacute stroke outcome. Healthy young adult mice were subjected to transient 30 min middle cerebral artery occlusion (MCAo)/reperfusion. Daily treatment with aleglitazar was begun on the day of MCAo and continued until sacrifice. Blood glucose measurements and lipid profile did not differ between mice receiving aleglitazar and mice receiving vehicle after MCAo. Aleglitazar reduced the size of the ischemic lesion as assessed using NeuN immunohistochemistry on day 7. Sensorimotor performance on the rotarod was impaired during the first week after MCAo, an effect that was significantly attenuated by treatment with aleglitazar. Smaller lesion volume in mice treated with aleglitazar was accompanied by a decrease in mRNA transcription of IL-1ß, Vcam-1, and Icam-1, suggesting that reduced proinflammatory signaling and reduced vascular inflammation in the ischemic hemisphere contribute to the beneficial effects of aleglitazar during the first week after stroke. Further experiments in primary murine microglia confirmed that aleglitazar reduces key aspects of microglia activation including NO production, release of proinflammatory cytokines, migration, and phagocytosis. In aggregate, a brief course of PPARα/γ agonist aleglitazar initiated post-event affords stroke protection and functional recovery in a model of mild brain ischemia. Our data underscores the theme of delayed injury processes such as neuroinflammation as promising therapeutic targets in stroke. KEY MESSAGES: PPARα/γ agonist aleglitazar improves stroke outcome after transient brain ischemia. Aleglitazar attenuates inflammatory responses in post-ischemic brain. Aleglitazar reduces microglia migration, phagocytosis, and release of cytokines. Beneficial effects of aleglitazar independent of glucose regulation. Aleglitazar provides extended window of opportunity for stroke treatment.


Subject(s)
Brain Ischemia , Oxazoles/pharmacology , PPAR alpha/agonists , PPAR gamma/agonists , Stroke , Thiophenes/pharmacology , Animals , Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain Ischemia/prevention & control , Disease Models, Animal , Mice , PPAR alpha/metabolism , PPAR gamma/metabolism , Stroke/metabolism , Stroke/pathology , Stroke/prevention & control
5.
Exp Neurol ; 306: 34-44, 2018 08.
Article in English | MEDLINE | ID: mdl-29684438

ABSTRACT

Evidence for a critical pathophysiological role of aberrant cytoskeletal dynamics is being uncovered in a growing number of neuropsychiatric syndromes. A sedentary lifestyle as well as overt psychopathology is prevalent in patients with the metabolic syndrome. Using mice deficient in gelsolin (Gsn-/-), a crucial actin-severing protein, we here investigated reduced actin turnover as a potential common driver of metabolic disturbances, sedentary behavior, and an anxious/depressive phenotype. Gelsolin deficiency resulted in reduced lifespan. As compared to wildtype controls, Gsn-/- mice (~ 9 weeks) fed a high-fat diet (HFD) over a span of 12 weeks showed increased body weight gain, fat mass, hepatic steatosis, and adipocyte hypertrophy as well as a significantly reduced respiratory quotient. Moreover, increased rigidity of the actin cytoskeleton in mice on HFD induced mRNA expression of Acc1, Acc2, Fasn, and Lipe, key genes involved in fatty acid metabolism in the liver. Glucose tolerance and insulin sensitivity were worsened in Gsn-/- HFD relative to Gsn+/+ HFD mice. Hypertension in Gsn-/- mice was associated with reduced endothelial NO synthase (eNOS) mRNA expression and reduced eNOS protein trafficking to the plasma membrane. Furthermore, acetylcholine-induced cGMP production and relaxation of aortic rings were impaired by actin filament stabilization. Gsn-/- mice on HFD displayed reduced corticosterone concentrations and reduced energy expenditure as compared to Gsn+/+ HFD mice. Moreover, Gsn-/- HFD mice displayed an overall pattern of hypoactive and anxious/depressive-like behavior. In aggregate, our results demonstrate that impaired actin filament dynamics promote the development of key behavioral and physiological aspects of the metabolic syndrome.


Subject(s)
Actin Cytoskeleton/metabolism , Cytoskeleton/metabolism , Cytoskeleton/pathology , Sedentary Behavior , Adipocytes/pathology , Animals , Behavior, Animal , Diet, High-Fat/adverse effects , Disease Models, Animal , Gelsolin/deficiency , Gelsolin/genetics , Gene Expression Regulation , Hypertension/etiology , Hypertension/physiopathology , Liver/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Obesity/complications , Obesity/genetics , Obesity/pathology , Weight Gain
6.
Acta Neuropathol ; 135(4): 551-568, 2018 04.
Article in English | MEDLINE | ID: mdl-29249001

ABSTRACT

After stroke, macrophages in the ischemic brain may be derived from either resident microglia or infiltrating monocytes. Using bone marrow (BM)-chimerism and dual-reporter transgenic fate mapping, we here set out to delimit the responses of either cell type to mild brain ischemia in a mouse model of 30 min transient middle cerebral artery occlusion (MCAo). A discriminatory analysis of gene expression at 7 days post-event yielded 472 transcripts predominantly or exclusively expressed in blood-derived macrophages as well as 970 transcripts for microglia. The differentially regulated genes were further collated with oligodendrocyte, astrocyte, and neuron transcriptomes, resulting in a dataset of microglia- and monocyte-specific genes in the ischemic brain. Functional categories significantly enriched in monocytes included migration, proliferation, and calcium signaling, indicative of strong activation. Whole-cell patch-clamp analysis further confirmed this highly activated state by demonstrating delayed outward K+ currents selectively in invading cells. Although both cell types displayed a mixture of known phenotypes pointing to the significance of 'intermediate states' in vivo, blood-derived macrophages were generally more skewed toward an M2 neuroprotective phenotype. Finally, we found that decreased engraftment of blood-borne cells in the ischemic brain of chimeras reconstituted with BM from Selplg-/- mice resulted in increased lesions at 7 days and worse post-stroke sensorimotor performance. In aggregate, our study establishes crucial differences in activation state between resident microglia and invading macrophages after stroke and identifies unique genomic signatures for either cell type.


Subject(s)
Brain Ischemia/metabolism , Macrophages/metabolism , Microglia/metabolism , Stroke/metabolism , Animals , Brain/metabolism , Brain/pathology , Brain Ischemia/pathology , Cations, Monovalent/metabolism , Disease Models, Animal , Gene Expression , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Macrophages/pathology , Male , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Membrane Potentials/physiology , Mice, Inbred C57BL , Mice, Transgenic , Microglia/pathology , Potassium/metabolism , Stroke/pathology , Transplantation Chimera
7.
Eur Arch Psychiatry Clin Neurosci ; 267(5): 473-477, 2017 Aug.
Article in English | MEDLINE | ID: mdl-27896432

ABSTRACT

Microglia senescence may promote neuropsychiatric disease. This prompted us to examine the relationship between microglia activation states and telomere biology. A panel of candidate genes associated with telomere maintenance, mitochondrial biogenesis, and cell-cycle regulation were investigated in M1- and M2-polarized microglia in vitro as well as in MACS-purified CD11b+ microglia/brain macrophages from models of stroke, Alzheimer's disease, and chronic stress. M1 polarization, ischemia, and Alzheimer pathology elicited a strikingly similar transcriptomic profile with, in particular, reduced expression of murine Tert. Our results link classical microglia activation with repression of telomere-associated genes, suggesting a new mechanism underlying microglia dysfunction.


Subject(s)
Gene Expression Regulation/genetics , Infarction, Middle Cerebral Artery/pathology , Microglia/metabolism , Telomerase/metabolism , Animals , CD11b Antigen/metabolism , Cell Polarity/drug effects , Cell Polarity/genetics , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Gene Expression Regulation/drug effects , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Mutation/genetics , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Presenilin-1/genetics , RNA, Messenger/metabolism , Telomerase/genetics
8.
BMC Neurol ; 16(1): 144, 2016 Aug 18.
Article in English | MEDLINE | ID: mdl-27538712

ABSTRACT

BACKGROUND: VE-cadherin is the chief constituent of endothelial adherens junctions. However, the role of VE-cadherin in the pathogenesis of cerebrovascular diseases including brain ischemia has not yet been investigated. METHODS: VE-cadherin heterozygous (VEC(+/-)) mice and wildtype controls were subjected to transient brain ischemia by 30 min filamentous middle cerebral artery occlusion (MCAo)/reperfusion. RESULTS: Acute lesion sizes as assessed by MR-imaging on day 3 did not differ between genotypes. Unexpectedly, however, partial loss of VE-cadherin resulted in long-term stroke protection measured histologically on day 28. Equally surprisingly, VEC(+/-) mice displayed no differences in post-stroke angiogenesis compared to littermate controls, but showed increased absolute regional cerebral blood flow in ischemic striatum at four weeks. The early induction of VE-cadherin mRNA transcription after stroke was reduced in VEC(+/-) mice. By contrast, N-cadherin and ß-catenin mRNA expression showed a delayed, but sustained, upregulation up to 28 days after MCAo, which was increased in VEC(+/-) mice. Furthermore, partial loss of VE-cadherin resulted in a pattern of elevated ischemia-triggered mRNA transcription of pericyte-related molecules α-smooth muscle actin (α-SMA), aminopeptidase N (CD13), and platelet-derived growth factor receptor ß (PDGFR-ß). CONCLUSIONS: Partial loss of VE-cadherin results in long term stroke protection. On the cellular and molecular level, this effect appears to be mediated by improved endothelial/pericyte interactions and the resultant increase in cerebral blood flow. Our study reinforces accumulating evidence that long-term stroke outcome depends critically on vascular mechanisms.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Cerebrovascular Circulation/physiology , Endothelium, Vascular/metabolism , Infarction, Middle Cerebral Artery/metabolism , Ischemic Attack, Transient/metabolism , Pericytes/metabolism , Animals , Disease Models, Animal , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/physiopathology , Ischemic Attack, Transient/etiology , Ischemic Attack, Transient/physiopathology , Male , Mice , Mice, Transgenic
9.
Brain Struct Funct ; 221(5): 2717-34, 2016 06.
Article in English | MEDLINE | ID: mdl-25989853

ABSTRACT

Impaired actin filament dynamics have been associated with cellular senescence. Microglia, the resident immune cells of the brain, are emerging as a central pathophysiological player in neurodegeneration. Microglia activation, which ranges on a continuum between classical and alternative, may be of critical importance to brain disease. Using genetic and pharmacological manipulations, we studied the effects of alterations in actin dynamics on microglia effector functions. Disruption of actin dynamics did not affect transcription of genes involved in the LPS-triggered classical inflammatory response. By contrast, in consequence of impaired nuclear translocation of phospho-STAT6, genes involved in IL-4 induced alternative activation were strongly downregulated. Functionally, impaired actin dynamics resulted in reduced NO secretion and reduced release of TNFalpha and IL-6 from LPS-stimulated microglia and of IGF-1 from IL-4 stimulated microglia. However, pathological stabilization of the actin cytoskeleton increased LPS-induced release of IL-1beta and IL-18, which belong to an unconventional secretory pathway. Reduced NO release was associated with decreased cytoplasmic iNOS protein expression and decreased intracellular arginine uptake. Furthermore, disruption of actin dynamics resulted in reduced microglia migration, proliferation and phagocytosis. Finally, baseline and ATP-induced [Ca(2+)]int levels were significantly increased in microglia lacking gelsolin, a key actin-severing protein. Together, the dynamic state of the actin cytoskeleton profoundly and distinctly affects microglia behaviours. Disruption of actin dynamics attenuates M2 polarization by inhibiting transcription of alternative activation genes. In classical activation, the role of actin remodelling is complex, does not relate to gene transcription and shows a major divergence between cytokines following conventional and unconventional secretion.


Subject(s)
Actins/metabolism , Microglia/metabolism , Animals , Calcium/metabolism , Cell Proliferation , Cells, Cultured , Chemotaxis , Cytokines/metabolism , Encephalitis/chemically induced , Encephalitis/metabolism , Gelsolin/genetics , Interleukin-4/metabolism , Lipopolysaccharides , Mice , Mice, Knockout , Nitrogen Oxides/metabolism , Phagocytosis , STAT6 Transcription Factor/metabolism
10.
J Neural Transm (Vienna) ; 122(9): 1329-38, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25981674

ABSTRACT

Recent evidence from animal and human studies suggests neuroprotective effects of the SSRI fluoxetine, e.g., in the aftermath of stroke. The underlying molecular mechanisms remain to be fully defined. Because of its effects on the cytochrome P450 system (CYP450), we hypothesized that neuroprotection by fluoxetine is related to altered metabolism of retinoic acid (RA), whose CYP450-mediated degradation in brain tissue constitutes an important step in the regulation of its site-specific auto- and paracrine actions. Using traditional pharmacological in vitro assays, the effects of fluoxetine on RA degradation were probed in crude synaptosomes from rat brain and human-derived SH-SY5Y cells, and in cultures of neuron-like SH-SY5Y cells. Furthermore, retinoid-dependent effects of fluoxetine on neuronal survival following glutamate exposure were investigated in rat primary neurons cells using specific retinoid receptor antagonists. Experiments revealed dose-dependent inhibition of synaptosomal RA degradation by fluoxetine along with dose-dependent increases in RA levels in cell cultures. Furthermore, fluoxetine's neuroprotective effects against glutamate excitotoxicity in rat primary neurons were demonstrated to partially depend on RA signaling. Taken together, these findings demonstrate for the first time that the potent, pleiotropic antidepressant fluoxetine directly interacts with RA homeostasis in brain tissue, thereby exerting its neuroprotective effects.


Subject(s)
Cerebral Cortex/drug effects , Fluoxetine/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Tretinoin/metabolism , Animals , Antidepressive Agents, Second-Generation/pharmacology , Benzhydryl Compounds/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Cerebral Cortex/physiology , Chromatography, High Pressure Liquid , Diazepam/pharmacology , Dose-Response Relationship, Drug , Glutamic Acid/metabolism , Glutamic Acid/toxicity , Humans , Modafinil , Neurons/physiology , Rats, Wistar , Receptors, Retinoic Acid/antagonists & inhibitors , Receptors, Retinoic Acid/metabolism , Synaptosomes/drug effects , Synaptosomes/metabolism
11.
J Neuroimmunol ; 256(1-2): 1-6, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23385081

ABSTRACT

In the brain, retinoic acid (RA) concentrations are under tight spatio-temporal control. Here, we show that challenge of primary mouse microglia with lipopolysaccharide (LPS) results in increased release of nitric oxide (NO) and tumor necrosis factor-α (TNF-α). Co-administration of RA attenuated microglial activation. Similarly, pretreatment with RA-metabolism inhibitor liarozole potently reduced NO and TNF-α release. Conversely, activated microglia showed increased protein expression of RA-degrading cytochromes CYP26A1, CYP26B1, CYP3A4 and CYP2C. Correspondingly, RA catabolism by activated microglia was significantly increased. Our results indicate that RA reduces microglial activation, but also, conversely, that the activation state of microglia influences RA metabolism.


Subject(s)
Gene Expression Regulation/physiology , Microglia/metabolism , Tretinoin/metabolism , Analysis of Variance , Animals , Animals, Newborn , Brain/cytology , Cells, Cultured , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Imidazoles/pharmacology , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Microglia/drug effects , Nitric Oxide/metabolism , RNA, Messenger/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Retinoid X Receptors/genetics , Retinoid X Receptors/metabolism , Time Factors , Tretinoin/pharmacology , Tumor Necrosis Factor-alpha/metabolism
12.
Eur Arch Psychiatry Clin Neurosci ; 262(8): 697-704, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22434147

ABSTRACT

While the biological importance of the cytochrome P450 system in the liver is well established, much less is known about its role in the brain and drug interactions at the level of brain cells have hardly been investigated. Here, we show that modafinil, a well-known inducer of hepatic CYP enzymes, also increases CYP3A4 expression in human-derived neuron-like SH-SY5Y cells. Upregulation of CYP3A4 by modafinil was associated with increased retinoic acid (RA) degradation, which could be blocked by specific CYP3A4 inhibitor erythromycin. In turn, reduced RA levels in culture medium during modafinil treatment resulted in decreased neuronal differentiation of SH-SY5Y cells as assessed by intracellular neurotransmitter concentrations and proliferative activity. Again, this differentiation-impeding effect of modafinil on SH-SY5Y cells was antagonized by erythromycin. Similarly, modafinil treatment of the murine GL261 glioma cell line resulted in increased proliferative activity. This was associated with upregulation of RA-degrading CYP26A1 in GL261 cells. Taken together, our results indicate that psychopharmacological agents such as modafinil may directly act on CYP enzymes in neural tissue. These kinds of drug effects may become highly relevant especially in the context of biomolecules such as RA whose local metabolism in brain is under tight spatial and temporal control.


Subject(s)
Antineoplastic Agents/pharmacology , Benzhydryl Compounds/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Tretinoin/pharmacology , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Cell Differentiation/drug effects , Cell Line, Tumor , Cytochrome P-450 CYP3A/metabolism , Dopamine/metabolism , Erythromycin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Green Fluorescent Proteins/genetics , Humans , Mice , Modafinil , Neuroblastoma/pathology , Neurofilament Proteins/metabolism , Phosphopyruvate Hydratase/metabolism , Protein Synthesis Inhibitors/pharmacology , Transfection
13.
Plant J ; 58(2): 333-46, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19143996

ABSTRACT

Leaf senescence, the final step of leaf development, involves extensive reprogramming of gene expression. Here, we show that these processes include discrete changes of epigenetic indexing, as well as global alterations in chromatin organization. During leaf senescence, the interphase nuclei show a decondensation of chromocenter heterochromatin, and changes in the nuclear distribution of the H3K4me2, H3K4me3, and the H3K27me2 and H3K27me3 histone modification marks that index active and inactive chromatin, respectively. Locus-specific epigenetic indexing was studied at the WRKY53 key regulator of leaf senescence. During senescence, when the locus becomes activated, H3K4me2 and H3K4me3 are significantly increased at the 5' end and at coding regions. Impairment of these processes is observed in plants overexpressing the SUVH2 histone methyltransferase, which causes ectopic heterochromatization. In these plants the transcriptional initiation of WRKY53 and of the senescence-associated genes SIRK, SAG101, ANAC083, SAG12 and SAG24 is inhibited, resulting in a delay of leaf senescence. In SUVH2 overexpression plants, significant levels of H3K27me2 and H3K27me3 are detected at the 5'-end region of WRKY53, resulting in its transcriptional repression. Furthermore, SUVH2 overexpression inhibits senescence-associated global changes in chromatin organization. Our data suggest that complex epigenetic processes control the senescence-specific gene expression pattern.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/genetics , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Histones/metabolism , Plant Leaves/growth & development , Arabidopsis/growth & development , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Chromatin/metabolism , DNA-Binding Proteins/genetics , Gene Expression Regulation, Plant , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Methylation , RNA, Plant/genetics , Transcriptional Activation
14.
Plant Mol Biol ; 69(1-2): 213-26, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18974936

ABSTRACT

HIPP26 from Arabidopsis thaliana belongs to a novel class of plant proteins, characterized by a heavy metal associated domain and an additional isoprenylation motif. It is induced during cold, salt and drought stress. The nuclear localization of HIPP26, predicted by a NLS motif, could be confirmed in onion epidermal cells overexpressing GFP-HIPP26. Experiments with modified HIPP26 indicate that the isoprenylation plays a role in the spatial distribution in the nucleus. Using promoter-GUS constructs, a tissue specific expression pattern of HIPP26 could be shown, with high expression in the vascular tissue. By a yeast-two-hybrid approach a strong interaction of HIPP26 with the zinc finger homeodomain transcription factor ATHB29, which is known to play a role in dehydration stress response could be detected. This was confirmed by GST pull-down assays. When using a modified HIPP26 lacking the two central cysteines of the heavy metal associated domain, ATHB29 was not bound in the GST pull-down assay, indicating that this structure is necessary for the interaction. Further yeast-two-hybrid analyses testing interaction of different members of the HIPP family with related zinc finger transcription factors revealed a specific interaction of ATHB29 with several HIPP proteins. A functional relationship between HIPP26 and ATHB29 is also indicated by experiments with mutants of HIPP26 showing altered expression levels of such genes known to be regulated by ATHB29.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Cell Nucleus/metabolism , Droughts , Metals, Heavy/metabolism , Transcription Factors/metabolism , Zinc Fingers , Phylogeny , Protein Binding , Protein Prenylation , Reverse Transcriptase Polymerase Chain Reaction , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...