Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Antimicrob Agents Chemother ; 59(4): 2016-28, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25605356

ABSTRACT

New agents are urgently needed for the therapeutic treatment of Staphylococcus aureus infections. In that regard, S. aureus RNase RnpA may represent a promising novel dual-function antimicrobial target that participates in two essential cellular processes, RNA degradation and tRNA maturation. Accordingly, we previously used a high-throughput screen to identify small-molecule inhibitors of the RNA-degrading activity of the enzyme and showed that the RnpA inhibitor RNPA1000 is an attractive antimicrobial development candidate. In this study, we used a series of in vitro and cellular assays to characterize a second RnpA inhibitor, RNPA2000, which was identified in our initial screening campaign and is structurally distinct from RNPA1000. In doing so, it was found that S. aureus RnpA does indeed participate in 5'-precursor tRNA processing, as was previously hypothesized. Further, we show that RNPA2000 is a bactericidal agent that inhibits both RnpA-associated RNA degradation and tRNA maturation activities both in vitro and within S. aureus. The compound appears to display specificity for RnpA, as it did not significantly affect the in vitro activities of unrelated bacterial or eukaryotic ribonucleases and did not display measurable human cytotoxicity. Finally, we show that RNPA2000 exhibits antimicrobial activity and inhibits tRNA processing in efflux-deficient Gram-negative pathogens. Taken together, these data support the targeting of RnpA for antimicrobial development purposes, establish that small-molecule inhibitors of both of the functions of the enzyme can be identified, and lend evidence that RnpA inhibitors may have broad-spectrum antimicrobial activities.


Subject(s)
Anti-Bacterial Agents/pharmacology , RNA, Bacterial/drug effects , RNA, Transfer/drug effects , Ribonuclease P/antagonists & inhibitors , Staphylococcus aureus/drug effects , Staphylococcus aureus/metabolism , Cell Line , Cell Survival/drug effects , High-Throughput Screening Assays , Humans , Hydrazines/pharmacology , Microbial Sensitivity Tests , Small Molecule Libraries , Thiourea/analogs & derivatives , Thiourea/pharmacology , Transcription, Genetic/drug effects
2.
Cell ; 159(2): 281-94, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25303525

ABSTRACT

Activity-dependent CREB phosphorylation and gene expression are critical for long-term neuronal plasticity. Local signaling at CaV1 channels triggers these events, but how information is relayed onward to the nucleus remains unclear. Here, we report a mechanism that mediates long-distance communication within cells: a shuttle that transports Ca(2+)/calmodulin from the surface membrane to the nucleus. We show that the shuttle protein is γCaMKII, its phosphorylation at Thr287 by ßCaMKII protects the Ca(2+)/CaM signal, and CaN triggers its nuclear translocation. Both ßCaMKII and CaN act in close proximity to CaV1 channels, supporting their dominance, whereas γCaMKII operates as a carrier, not as a kinase. Upon arrival within the nucleus, Ca(2+)/CaM activates CaMKK and its substrate CaMKIV, the CREB kinase. This mechanism resolves long-standing puzzles about CaM/CaMK-dependent signaling to the nucleus. The significance of the mechanism is emphasized by dysregulation of CaV1, γCaMKII, ßCaMKII, and CaN in multiple neuropsychiatric disorders.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Animals , Calcium/metabolism , Calcium Channels/metabolism , Calmodulin/metabolism , Cell Nucleus/metabolism , Neurons/metabolism , Phosphorylation , Rats, Sprague-Dawley , Transcription, Genetic
3.
FEBS Lett ; 587(6): 756-62, 2013 Mar 18.
Article in English | MEDLINE | ID: mdl-23395614

ABSTRACT

Recent studies suggest a link between mitochondria and proinflammatory cytokine generation. We previously demonstrated that overexpression of mitochondrial chaperone glucose-regulated protein75 (Grp75/mortalin) protects mitochondria. In this study we investigated the modulation of the lipopolisaccharide (LPS)-induced inflammatory response of microglial BV-2 cells by Grp75. We demonstrate that LPS-induced activation promotes significant metabolic changes suppressing mitochondrial function and increasing glycolysis. Overexpression of Grp75 attenuates the LPS-induced oxidative and metabolic responses, and suppresses proinflammatory activation, which depends on both NF-κB activation and lactate. Thus overexpression of Grp75 provides a novel strategy to modulate proinflammatory cytokine production of relevance to inflammation-associated pathologies.


Subject(s)
HSP70 Heat-Shock Proteins/genetics , Membrane Proteins/genetics , Microglia/metabolism , Mitochondria/metabolism , Molecular Chaperones/genetics , Animals , Cells, Cultured , Cytokines/biosynthesis , Gene Expression Regulation/drug effects , Glycolysis/drug effects , Glycolysis/genetics , HSP70 Heat-Shock Proteins/metabolism , Inflammation/metabolism , Inflammation/pathology , Lactic Acid/metabolism , Lipopolysaccharides/pharmacology , Membrane Proteins/metabolism , Mice , Microglia/drug effects , Microglia/pathology , Mitochondria/drug effects , Mitochondria/genetics , Molecular Chaperones/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Oxidative Phosphorylation/drug effects , Plasmids , Transfection
4.
BMC Bioinformatics ; 12: 276, 2011 Jul 05.
Article in English | MEDLINE | ID: mdl-21729324

ABSTRACT

BACKGROUND: Activation of the NF-κB transcription factor and its associated gene expression in microglia is a key component in the response to brain injury. Its activation is dynamic and is part of a network of biochemical species with multiple feedback regulatory mechanisms. Mathematical modeling, which has been instrumental for understanding the NF-κB response in other cell types, offers a valuable tool to investigate the regulation of NF-κB activation in microglia at a systems level. RESULTS: We quantify the dynamic response of NF-κB activation and activation of the upstream kinase IKK using ELISA measurements of a microglial cell line following treatment with the pro-inflammatory cytokine TNFα. A new mathematical model is developed based on these data sets using a modular procedure that exploits the feedback structure of the network. We show that the new model requires previously unmodeled dynamics involved in the stimulus-induced degradation of the inhibitor IκBα in order to properly describe microglial NF-κB activation in a statistically consistent manner. This suggests a more prominent role for the ubiquitin-proteasome system in regulating the activation of NF-κB to inflammatory stimuli. We also find that the introduction of nonlinearities in the kinetics of IKK activation and inactivation is essential for proper characterization of transient IKK activity and corresponds to known biological mechanisms. Numerical analyses of the model highlight key regulators of the microglial NF-κB response, as well as those governing IKK activation. Results illustrate the dynamic regulatory mechanisms and the robust yet fragile nature of the negative feedback regulated network. CONCLUSIONS: We have developed a new mathematical model that incorporates previously unmodeled dynamics to characterize the dynamic response of the NF-κB signaling network in microglia. This model is the first of its kind for microglia and provides a tool for the quantitative, systems level study the dynamic cellular response to inflammatory stimuli.


Subject(s)
Brain Ischemia/metabolism , Microglia/metabolism , NF-kappa B/metabolism , Signal Transduction , Animals , Brain Ischemia/pathology , Cell Line , Cytokines/metabolism , Gene Expression Regulation , I-kappa B Kinase/metabolism , Inflammation/metabolism , Mice , NF-kappa B/antagonists & inhibitors , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism
5.
Mitochondrion ; 11(2): 279-86, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21047562

ABSTRACT

Ca(2+) transfer from endoplasmic reticulum (ER) to mitochondria at contact sites between the organelles can induce mitochondrial dysfunction and programmed cell death after stress. The ER-localized chaperone glucose-regulated protein 78kDa (GRP78/BiP) protects neurons against excitotoxicity and apoptosis. Here we show that overexpressing GRP78 protects astrocytes against ischemic injury, reduces net flux of Ca(2+) from ER to mitochondria, increases Ca(2+) uptake capacity in isolated mitochondria, reduces free radical production, and preserves respiratory activity and mitochondrial membrane potential after stress. We conclude that GRP78 influences ER-mitochondrial Ca(2+) crosstalk to maintain mitochondrial function and protect astrocytes from ischemic injury.


Subject(s)
Astrocytes/metabolism , Calcium/metabolism , Heat-Shock Proteins/metabolism , Ischemia/metabolism , Mitochondria/metabolism , Animals , Cells, Cultured , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Immunohistochemistry , Ischemia/physiopathology , Membrane Potentials , Mice , Mitochondria/physiology , Reactive Oxygen Species/metabolism
6.
J Neurosci ; 30(37): 12242-51, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20844120

ABSTRACT

The impairment of hippocampal neurogenesis has been linked to the pathogenesis of neurological disorders from chronic neurodegenerative disease to the progressive cognitive impairment of children who receive brain irradiation. Numerous studies provide evidence that inflammation downregulates neurogenesis, with multiple factors contributing to this impairment. Although mitochondria are one of the primary targets of inflammatory injury, the role of mitochondrial function in the modulation of neurogenesis remains relatively unstudied. In this study, we used neurosphere-derived cells to show that immature doublecortin (Dcx)-positive neurons are uniquely sensitive to mitochondrial inhibition, demonstrating rapid loss of mitochondrial potential and cell viability compared with glial cells and more mature neurons. Mitochondrial inhibition for 24 h produced no significant changes in astrocyte or oligodendrocyte viability, but reduced viability of mature neurons by 30%, and reduced survival of Dcx(+) cells by 60%. We demonstrate that protection of mitochondrial function with mitochondrial metabolites or the mitochondrial chaperone mtHsp75/mortalin partially reverses the inflammation-associated impairment of neurogenesis in vitro and in irradiated mice in vivo. Our findings highlight mitochondrial mechanisms involved in neurogenesis and indicate mitochondria as a potential target for protective strategies to prevent the impairment of neurogenesis by inflammation.


Subject(s)
Energy Metabolism/physiology , Inflammation Mediators/physiology , Mitochondria/pathology , Neural Inhibition/physiology , Neurogenesis/physiology , Animals , Cell Differentiation/genetics , Cell Line , Cells, Cultured , Doublecortin Domain Proteins , Doublecortin Protein , Energy Metabolism/genetics , HSP70 Heat-Shock Proteins/biosynthesis , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/physiology , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/biosynthesis , Mitochondria/genetics , Mitochondria/physiology , Neural Inhibition/genetics , Neurogenesis/genetics , Neurons/metabolism , Neurons/pathology , Neuropeptides/antagonists & inhibitors , Neuropeptides/biosynthesis , Thiamine/administration & dosage
7.
Glia ; 58(9): 1042-9, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20235222

ABSTRACT

Brief forebrain ischemia is a model of the delayed hippocampal neuronal loss seen in patients following cardiac arrest and resuscitation. Previous studies demonstrated that selective dysfunction of hippocampal CA1 subregion astrocytes occurs hours to days before delayed neuronal death. In this study we tested the strategy of directing protection to astrocytes to protect neighboring neurons from forebrain ischemia. Two well-studied protective proteins, heat shock protein 72 (Hsp72) or superoxide dismutase 2 (SOD2), were genetically targeted for expression in astrocytes using the astrocyte-specific human glial fibrillary acidic protein (GFAP) promoter. The expression constructs were injected stereotacticly immediately above the hippocampal CA1 region on one side of the rat brain two days prior to forebrain ischemia. Cell type specific expression was confirmed by double label immunohistochemistry. When the expression constructs were injected two days before transient forebrain ischemia, the loss of CA1 hippocampal neurons observed seven days later was significantly reduced on the injected side compared with controls. This neuroprotection was associated with significantly better preservation of astrocyte glutamate transporter-1 immunoreactivity at 5-h reperfusion and reduced oxidative stress. Improving the resistance of astrocytes to ischemic stress by targeting either the cytosolic or mitochondrial compartment was thus associated with preservation of CA1 neurons following forebrain ischemia. Targeting astrocytes is a promising strategy for neuronal preservation following cardiac arrest and resuscitation.


Subject(s)
Astrocytes/physiology , Brain Ischemia/physiopathology , CA1 Region, Hippocampal/physiopathology , HSP72 Heat-Shock Proteins/metabolism , Neurons/physiology , Superoxide Dismutase/metabolism , Animals , CA1 Region, Hippocampal/pathology , Cell Hypoxia/physiology , Cells, Cultured , Excitatory Amino Acid Transporter 2/metabolism , Glial Fibrillary Acidic Protein/genetics , HSP72 Heat-Shock Proteins/genetics , Humans , Male , Mice , Neurons/pathology , Oxidative Stress/physiology , Promoter Regions, Genetic , Prosencephalon/pathology , Prosencephalon/physiopathology , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/genetics , Transfection
8.
J Cereb Blood Flow Metab ; 29(2): 365-74, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18985056

ABSTRACT

Mitochondria are known to be central to the cell's response to ischemia, because of their role in energy generation, in free radical generation, and in the regulation of apoptosis. Heat shock protein 75 (Hsp75/Grp75/mortalin/TRAP1) is a member of the HSP70 chaperone family, which is targeted to mitochondria. Overexpression of Hsp75 was achieved in rat brain by DNA transfection, and expression was observed in both astrocytes and neurons. Rats were subjected to 100 mins middle cerebral artery occlusion followed by assessment of infarct volume, neurological score, mitochondrial function, and levels of oxidative stress at 24 h reperfusion. Overexpression of Hsp75 reduced infarct area from 44.6%+/-21.1% to 25.7%+/-12.1% and improved neurological outcome significantly. This was associated with improved mitochondrial function as shown by protection of complex IV activity, marked reduction of free radical generation detected by hydroethidine fluorescence, reduction of lipid peroxidation detected by 4-hydroxy-2-nonenol immunoreactivity, and increased preservation of ATP levels. This suggests that targeting mitochondria for protection may be a useful strategy to reduce ischemic brain injury.


Subject(s)
Brain Ischemia/metabolism , Brain Ischemia/pathology , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Mitochondria/metabolism , Oxidative Stress , Adenosine Triphosphate/metabolism , Animals , Astrocytes/metabolism , Brain Ischemia/genetics , Electron Transport , Gene Expression Regulation , HSP70 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/genetics , Lipid Metabolism , Male , Neurons/metabolism , Rats , Rats, Sprague-Dawley
9.
Anesthesiology ; 109(2): 339-48, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18648242

ABSTRACT

Although heat shock proteins have been studied for decades, new intracellular and extracellular functions in a variety of diseases continue to be discovered. Heat shock proteins function within networks of interacting proteins; they can alter cellular physiology rapidly in response to stress without requiring new protein synthesis. This review focuses on the heat shock protein 70 family and considers especially the functions of the inducible member, heat shock protein 72, in the setting of cerebral ischemia. In general, inhibiting apoptotic signaling at multiple points and up-regulating survival signaling, heat shock protein 70 has a net prosurvival effect. Heat shock protein 70 has both antiinflammatory and proinflammatory effects depending on the cell type, context, and intracellular or extracellular location. Intracellular effects are often antiinflammatory with inhibition of nuclear factor-kappaB signaling. Extracellular effects can lead to inflammatory cytokine production or induction of regulatory immune cells and reduced inflammation.


Subject(s)
Apoptosis/physiology , Brain Ischemia/metabolism , HSP70 Heat-Shock Proteins/physiology , Inflammation/etiology , Animals , Cell Death/physiology , HSP70 Heat-Shock Proteins/metabolism , Humans , Inflammation/metabolism , Signal Transduction/physiology
10.
J Cereb Blood Flow Metab ; 28(5): 1009-16, 2008 May.
Article in English | MEDLINE | ID: mdl-18091755

ABSTRACT

Mitochondrial heat shock protein 70 (mtHsp70/Hsp75/Grp75/mortalin/TRAP-1/PBP74) is an essential mitochondrial chaperone and a member of the heat shock protein 70 (HSP70) family. Although many studies have shown the protective properties of overexpression of the cytosolic inducible member of the HSP70 family, Hsp72, few studies have investigated the protective potential of Hsp75 against ischemic injury. Mitochondria are one of the primary targets of ischemic injury in astrocytes. In this study, we analyzed the effects of Hsp75 overexpression on cellular levels of reactive oxygen species (ROS), mitochondrial membrane potential, ATP levels, and viability during the ischemia-like conditions of oxygen-glucose deprivation (OGD) or glucose deprivation (GD) in primary astrocytic cultures. We show that Hsp75 overexpression decreases ROS production and preserves mitochondrial membrane potential during GD, and preserves ATP levels and cell viability during OGD. These findings indicate that Hsp75 can provide protection against ischemia-like in vitro injury and suggest that it should be further studied as a potential candidate for protection against ischemic injury.


Subject(s)
Astrocytes/metabolism , Astrocytes/pathology , Brain Ischemia/metabolism , Brain Ischemia/pathology , HSP90 Heat-Shock Proteins/metabolism , Adenosine Triphosphate/metabolism , Animals , Carbamide Peroxide , Cell Survival/physiology , Cells, Cultured , Drug Combinations , Gene Expression/physiology , Glucose/pharmacology , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/genetics , Humans , In Vitro Techniques , Membrane Potential, Mitochondrial/physiology , Mice , Mitochondria/metabolism , Oxidative Stress/physiology , Peroxides/pharmacology , Transduction, Genetic , Urea/analogs & derivatives , Urea/pharmacology
11.
Curr Biol ; 13(20): 1768-74, 2003 Oct 14.
Article in English | MEDLINE | ID: mdl-14561401

ABSTRACT

BACKGROUND: Shoots of all land plants have a radial pattern that can be considered to have an adaxial (central)-abaxial (peripheral) polarity. In Arabidopsis, gain-of-function alleles of PHAVOLUTA and PHABULOSA, members of the class III HD-ZIP gene family, result in adaxialization of lateral organs. Conversely, loss-of-function alleles of the KANADI genes cause an adaxialization of lateral organs. Thus, the class III HD-ZIP and KANADI genes comprise a genetic system that patterns abaxial-adaxial polarity in lateral organs produced from the apical meristem. RESULTS: We show that gain-of-function alleles of REVOLUTA, another member of the class III HD-ZIP gene family, are characterized by adaxialized lateral organs and alterations in the radial patterning of vascular bundles in the stem. The gain-of-function phenotype can be obtained by changing only the REVOLUTA mRNA sequence and without changing the protein sequence; this finding indicates that this phenotype is likely mediated through an interference with microRNA binding. Loss of KANADI activity results in similar alterations in vascular patterning as compared to REVOLUTA gain-of-function alleles. Simultaneous loss-of-function of PHABULOSA, PHAVOLUTA, and REVOLUTA abaxializes cotyledons, abolishes the formation of the primary apical meristem, and in severe cases, eliminates bilateral symmetry; these phenotypes implicate these three genes in radial patterning of both embryonic and postembryonic growth. CONCLUSIONS: Based on complementary vascular and leaf phenotypes of class III HD-ZIP and KANADI mutants, we propose that a common genetic program dependent upon miRNAs governs adaxial-abaxial patterning of leaves and radial patterning of stems in the angiosperm shoot. This finding implies that a common patterning mechanism is shared between apical and vascular meristems.


Subject(s)
Arabidopsis Proteins/physiology , Arabidopsis/growth & development , Body Patterning/genetics , MicroRNAs/metabolism , Plant Shoots/growth & development , Transcription Factors/physiology , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Base Sequence , Body Patterning/physiology , Gene Expression Profiling , Models, Molecular , Molecular Sequence Data , Phylogeny , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...