Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
2.
ACS Chem Neurosci ; 14(2): 277-288, 2023 01 18.
Article in English | MEDLINE | ID: mdl-36574489

ABSTRACT

Age-dependent formation of insoluble protein aggregates is a hallmark of many neurodegenerative diseases. We are interested in the cell chemistry that drives the aggregation of polyQ-expanded mutant Huntingtin (mHtt) protein into insoluble inclusion bodies (IBs). Using an inducible cell model of Huntington's disease, we show that a transient cold shock (CS) at 4 °C followed by recovery incubation at temperatures of 25-37 °C strongly and rapidly induces the compaction of diffuse polyQ-expanded HuntingtinExon1-enhanced green fluorescent protein chimera protein (mHtt) into round, micron size, cytosolic IBs. This transient CS-induced mHtt IB formation is independent of microtubule integrity or de novo protein synthesis. The addition of millimolar concentrations of sodium chloride accelerates, whereas urea suppresses this transient CS-induced mHtt IB formation. These results suggest that the low temperature of CS constrains the conformation dynamics of the intrinsically disordered mHtt into labile intermediate structures to facilitate de-solvation and hydrophobic interaction for IB formation at the higher recovery temperature. This work, along with our previous observation of the effects of heat shock protein chaperones and osmolytes in driving mHtt IB formation, underscores the primacy of mHtt structuring and rigidification for H-bond-mediated cross-linking in a two-step mechanism of mHtt IB formation in living cells.


Subject(s)
Huntington Disease , Inclusion Bodies , Humans , Cold-Shock Response , Cytosol/metabolism , Huntingtin Protein/metabolism , Huntington Disease/genetics , Huntington Disease/metabolism , Inclusion Bodies/metabolism , Mutation/genetics
3.
Adv Exp Med Biol ; 1409: 23-49, 2023.
Article in English | MEDLINE | ID: mdl-35995906

ABSTRACT

Heat shock factor 1 (HSF1) is a master transcription regulator that mediates the induction of heat shock protein chaperones for quality control (QC) of the proteome and maintenance of proteostasis as a protective mechanism in response to stress. Research in this particular area has accelerated dramatically over the past three decades following successful isolation, cloning, and characterization of HSF1. The intricate multi-protein complexes and transcriptional activation orchestrated by HSF1 are fundamental processes within the cellular QC machinery. Our primary focus is on the regulation and function of HSF1 in aging and neurodegenerative diseases (ND) which represent physiological and pathological states of dysfunction in protein QC. This chapter presents an overview of HSF1 structural, functional, and energetic properties in healthy cells while addressing the deterioration of HSF1 function viz-à-viz age-dependent and neuron-specific vulnerability to ND. We discuss the structural domains of HSF1 with emphasis on the intrinsically disordered regions and note that disease proteins associated with ND are often structurally disordered and exquisitely sensitive to changes in cellular environment as may occur during aging. We propose a hypothesis that age-dependent changes of the intrinsically disordered proteome likely hold answers to understand many of the functional, structural, and organizational changes of proteins and signaling pathways in aging - dysfunction of HSF1 and accumulation of disease protein aggregates in ND included.Structured AbstractsIntroduction: Heat shock factor 1 (HSF1) is a master transcription regulator that mediates the induction of heat shock protein chaperones for quality control (QC) of the proteome as a cyto-protective mechanism in response to stress. There is cumulative evidence of age-related deterioration of this QC mechanism that contributes to disease vulnerability. OBJECTIVES: Herein we discuss the regulation and function of HSF1 as they relate to the pathophysiological changes of protein quality control in aging and neurodegenerative diseases (ND). METHODS: We present an overview of HSF1 structural, functional, and energetic properties in healthy cells while addressing the deterioration of HSF1 function vis-à-vis age-dependent and neuron-specific vulnerability to neurodegenerative diseases. RESULTS: We examine the impact of intrinsically disordered regions on the function of HSF1 and note that proteins associated with neurodegeneration are natively unstructured and exquisitely sensitive to changes in cellular environment as may occur during aging. CONCLUSIONS: We put forth a hypothesis that age-dependent changes of the intrinsically disordered proteome hold answers to understanding many of the functional, structural, and organizational changes of proteins - dysfunction of HSF1 in aging and appearance of disease protein aggregates in neurodegenerative diseases included.


Subject(s)
DNA-Binding Proteins , Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Heat Shock Transcription Factors/genetics , Heat Shock Transcription Factors/metabolism , Proteome/metabolism , Protein Aggregates , Heat-Shock Proteins , Molecular Chaperones/metabolism , Heat-Shock Response
4.
Stem Cell Reports ; 17(11): 2381-2391, 2022 11 08.
Article in English | MEDLINE | ID: mdl-36270284

ABSTRACT

Immunodeficient mice are widely used in human stem cell transplantation research. Recombination activating gene 1 (Rag1) deletion results in immunodeficiency and leads to accelerated aging in zebrafish with increased cytosolic accumulation of lipofuscin (LF). Unlike zebrafish, mammals have two homologs, Rag1 and Rag2, that regulate adaptive immunity. Currently, little is known if and how Rag1-/- and Rag2-/- may impact aging and LF accumulation in immunodeficient mouse brains and how this may confound results in human neural cell transplantation studies. Here, we demonstrate that in Rag2-/- mouse brains, LF appears early, spreads broadly, emits strong autofluorescence, and accumulates with age. LF is found in various types of glial cells, including xenografted human microglia. Surprisingly, in Rag1-/- mouse brains, LF autofluorescence is seen at much older ages compared with Rag2-/- brains. This study provides direct evidence that Rag2-/- expedites LF occurrence and sets a context for studies using aged immunodeficient mice.


Subject(s)
Lipofuscin , Animals , Humans , Mice , Brain/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Homeodomain Proteins/metabolism , Mammals/metabolism , Nuclear Proteins/metabolism , Stem Cell Transplantation
5.
Sci Rep ; 10(1): 15511, 2020 09 23.
Article in English | MEDLINE | ID: mdl-32968182

ABSTRACT

Osmolytes are organic solutes that change the protein folding landscape shifting the equilibrium towards the folded state. Herein, we use osmolytes to probe the structuring and aggregation of the intrinsically disordered mutant Huntingtin (mHtt) vis-a-vis the pathogenicity of mHtt on transcription factor function and cell survival. Using an inducible PC12 cell model of Huntington's disease (HD), we show that stabilizing polyol osmolytes drive the aggregation of Htt103QExon1-EGFP from a diffuse ensemble into inclusion bodies (IBs), whereas the destabilizing osmolyte urea does not. This effect of stabilizing osmolytes is innate, generic, countered by urea, and unaffected by HSP70 and HSC70 knockdown. A qualitatively similar result of osmolyte-induced mHtt IB formation is observed in a conditionally immortalized striatal neuron model of HD, and IB formation correlates with improved survival under stress. Increased expression of diffuse mHtt sequesters the CREB transcription factor to repress CREB-reporter gene activity. This repression is mitigated either by stabilizing osmolytes, which deplete diffuse mHtt or by urea, which negates protein-protein interaction. Our results show that stabilizing polyol osmolytes promote mHtt aggregation, alleviate CREB dysfunction, and promote survival under stress to support the hypothesis that lower molecular weight entities of disease protein are relevant pathogenic species in neurodegeneration.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Huntingtin Protein/metabolism , Huntington Disease/metabolism , Animals , Autophagy , Gene Knockdown Techniques , Glycerol/pharmacology , HSC70 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/metabolism , Huntingtin Protein/genetics , Mutation , Osmolar Concentration , PC12 Cells , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/metabolism , Protein Folding , Rats , Sorbitol/pharmacology , Sucrose/pharmacology , Trehalose/pharmacology
6.
J Biol Chem ; 293(40): 15581-15593, 2018 10 05.
Article in English | MEDLINE | ID: mdl-30143534

ABSTRACT

PolyQ-expanded huntingtin (mHtt) variants form aggregates, termed inclusion bodies (IBs), in individuals with and models of Huntington's disease (HD). The role of IB versus diffusible mHtt in neurotoxicity remains unclear. Using a ponasterone (PA)-inducible cell model of HD, here we evaluated the effects of heat shock on the appearance and functional outcome of Htt103QExon1-EGFP expression. Quantitative image analysis indicated that 80-90% of this mHtt protein initially appears as "diffuse" signals in the cytosol, with IBs forming at high mHtt expression. A 2-h heat shock during the PA induction reduced the diffuse signal, but greatly increased mHtt IB formation in both cytosol and nucleus. Dose- and time-dependent mHtt expression suggested that nucleated polymerization drives IB formation. RNA-mediated knockdown of heat shock protein 70 (HSP70) and heat shock cognate 70 protein (HSC70) provided evidence for their involvement in promoting diffuse mHtt to form IBs. Reporter gene assays assessing the impacts of diffuse versus IB mHtt showed concordance of diffuse mHtt expression with the repression of heat shock factor 1, cAMP-responsive element-binding protein (CREB), and NF-κB activity. CREB repression was reversed by heat shock coinciding with mHtt IB formation. In an embryonic striatal neuron-derived HD model, the chemical chaperone sorbitol similarly promoted the structuring of diffuse mHtt into IBs and supported cell survival under stress. Our results provide evidence that mHtt IB formation is a chaperone-supported cellular coping mechanism that depletes diffusible mHtt conformers, alleviates transcription factor dysfunction, and promotes neuron survival.


Subject(s)
Heat Shock Transcription Factors/genetics , Heat-Shock Response , Huntingtin Protein/genetics , Huntington Disease/genetics , Inclusion Bodies/metabolism , Neurons/metabolism , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Nucleus/pathology , Corpus Striatum/metabolism , Corpus Striatum/pathology , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Cytosol/drug effects , Cytosol/metabolism , Cytosol/pathology , Ecdysterone/analogs & derivatives , Ecdysterone/pharmacology , Embryo, Mammalian , Gene Expression Regulation , HSC70 Heat-Shock Proteins/genetics , HSC70 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Heat Shock Transcription Factors/metabolism , Huntingtin Protein/metabolism , Huntington Disease/chemically induced , Huntington Disease/metabolism , Huntington Disease/pathology , Inclusion Bodies/chemistry , Inclusion Bodies/drug effects , Models, Biological , Mutation , NF-kappa B/genetics , NF-kappa B/metabolism , Neurons/drug effects , Neurons/pathology , PC12 Cells , Primary Cell Culture , Rats , Sorbitol/pharmacology
7.
J Cell Physiol ; 229(9): 1224-35, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24435709

ABSTRACT

Neurons have a limited capacity for heat shock protein (HSP) induction and are vulnerable to the pathogenic consequence of protein misfolding and aggregation as seen in age-related neurodegenerative diseases. Sirtuin 1 (SIRT1), an NAD(+) -dependent lysine deacetylase with important biological functions, has been shown to sustain the DNA-binding state of HSF1 for HSP induction. Here we show that differentiation and maturation of embryonic cortical neurons and N2a neuroprogenitor cells is associated with decreases in SIRT1 expression and heat shock-dependent induction of HSP70 protein. Tests of a pharmacological activator and an inhibitor of SIRT1 affirm the regulatory role of SIRT1 in HSP70 induction. Protein cross-linking studies show that nuclear SIRT1 and HSF1 form a co-migrating high molecular weight complex upon stress. The use of retroviral vectors to manipulate SIRT1 expression in N2a cells show that shRNA-mediated knock down of SIRT1 causes spontaneous neurite outgrowth coincident with reduced growth rate and decreased induction of hsp70-reporter gene, whereas SIRT1 over-expression blocks the induced neural differentiation of N2a cells. Our results suggest that decreased SIRT1 expression is conducive to neuronal differentiation and this decrease contributes to the attenuated induction of HSPs in neurons.


Subject(s)
Cerebral Cortex/enzymology , Gene Knockdown Techniques , Heat-Shock Proteins/metabolism , Heat-Shock Response , Neural Stem Cells/enzymology , Neurogenesis , Neurons/enzymology , Sirtuin 1/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Cerebral Cortex/embryology , Cerebral Cortex/pathology , DNA-Binding Proteins/metabolism , Down-Regulation , Gestational Age , HSP70 Heat-Shock Proteins/metabolism , Heat Shock Transcription Factors , Heat-Shock Proteins/genetics , Mice , Neural Stem Cells/pathology , Neurons/pathology , Protein Binding , RNA Interference , Rats , Rats, Sprague-Dawley , Signal Transduction , Sirtuin 1/genetics , Time Factors , Transcription Factors/metabolism , Transfection
8.
Glia ; 61(3): 394-408, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23281078

ABSTRACT

Congenital hyperinsulinism/hyperammonemia (HI/HA) syndrome is caused by an activation mutation of glutamate dehydrogenase 1 (GDH1), a mitochondrial enzyme responsible for the reversible interconversion between glutamate and α-ketoglutarate. The syndrome presents clinically with hyperammonemia, significant episodic hypoglycemia, seizures, and frequent incidences of developmental and learning defects. Clinical research has implicated that although some of the developmental and neurological defects may be attributed to hypoglycemia, some characteristics cannot be ascribed to low glucose and as hyperammonemia is generally mild and asymptomatic, there exists the possibility that altered GDH1 activity within the brain leads to some clinical changes. GDH1 is allosterically regulated by many factors, and has been shown to be inhibited by the ADP-ribosyltransferase sirtuin 4 (SIRT4), a mitochondrially localized sirtuin. Here we show that SIRT4 is localized to mitochondria within the brain. SIRT4 is highly expressed in glial cells, specifically astrocytes, in the postnatal brain and in radial glia during embryogenesis. Furthermore, SIRT4 protein decreases in expression during development. We show that factors known to allosterically regulate GDH1 alter gliogenesis in CTX8 cells, a novel radial glial cell line. We find that SIRT4 and GDH1 overexpression play antagonistic roles in regulating gliogenesis and that a mutant variant of GDH1 found in HI/HA patients accelerates the development of glia from cultured radial glia cells.


Subject(s)
Cerebral Cortex/metabolism , Glutamate Dehydrogenase/metabolism , Neuroglia/metabolism , Sirtuins/metabolism , Animals , Cells, Cultured , Cerebral Cortex/cytology , Glutamate Dehydrogenase/genetics , Hyperammonemia/genetics , Hyperammonemia/metabolism , Hypoglycemia/genetics , Hypoglycemia/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Neuroglia/cytology , Rats , Sirtuins/genetics
9.
J Biol Chem ; 286(4): 2785-94, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21098017

ABSTRACT

Heat shock factor 1 (HSF1) mediates the cellular response to stress to increase the production of heat shock protein (HSP) chaperones for proper protein folding, trafficking, and degradation; failure of this homeostatic mechanism likely contributes to neurodegeneration. We show that the neuroprotective drug riluzole increased the amount of HSF1 in NG108-15 neuroprogenitor cells by slowing the specific turnover of HSF1 and supporting a more robust and sustained activation of HSF1. Using Hsp70-luciferase as a functional readout of the activity of HSF1, we show that riluzole amplified the heat shock induction of the reporter gene with an optimal increase at 1 µM. Immunocytochemical staining and Western blot quantitation of HSP70 in NG108-15 neuroprogenitor cells and embryonic spinal cord neurons provided corroborative evidence that riluzole amplified the HSF1-dependent regulation of HSP70 expression. Parallel studies on the GLT1 glutamate transporter showed that riluzole increased GLT1-reporter and GLT1 protein expression and that the increase was enhanced by heat shock and coincident with the increased expression of HSP70 and HSP90. This result is consistent with the anti-glutamatergic profile of riluzole and the presence of multiple heat shock elements on the GLT1 gene promoter, suggesting that riluzole may modulate GLT1 expression through HSF1. The increased HSP chaperones and GLT1 transporter blunted glutamate-induced and N-methyl D-aspartate receptor-mediated excitotoxic death. In summary, we show that riluzole increased the amount and activity of HSF1 to boost the expression of HSPs and GLT1 for neuroprotection under stress.


Subject(s)
DNA-Binding Proteins/metabolism , Excitatory Amino Acid Transporter 2/biosynthesis , Gene Expression Regulation/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Riluzole/pharmacology , Stem Cells/metabolism , Transcription Factors/metabolism , Animals , Cell Line , DNA-Binding Proteins/genetics , Excitatory Amino Acid Transporter 2/genetics , Glutamic Acid/metabolism , Glutamic Acid/pharmacology , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Heat Shock Transcription Factors , Heat-Shock Response/drug effects , Heat-Shock Response/genetics , Humans , Neurons/cytology , Rats , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Response Elements/physiology , Stem Cells/cytology , Transcription Factors/genetics
10.
J Cell Physiol ; 219(2): 485-93, 2009 May.
Article in English | MEDLINE | ID: mdl-19160416

ABSTRACT

Heat-induced cell death appears to be a cell-specific event. Chronic heat stress was lethal to human colon cancer cells (Caco-2, HT29, and HCT116), but not to normal diploid fibroblasts and other cancer cells (BJ-T, WI38, HeLa, ovarian 2008, WI38VA). Acute heat stress (45-51 degrees C, 30 min) caused cell death of colon cancer cells during recovery at physiological temperature. Thermal killing of Caco-2 cells was not mediated via oxidative stress since Caco-2 cells were much more resistant than HeLa and other cancer cells to H(2)O(2)-induced cell death. Acute heat stress caused a striking loss of eukaryotic initiation factor 5A (eIF5A) in colon cancer cells, but not in HeLa and other normal or transformed human fibroblasts. The heat-induced loss of eIF5A is likely to be due to changes in the protein stability. The half-life of eIF5A was changed from >20 h to less than 30 min during the acute heat stress. Sequence analysis of the eIF5A gene from Caco-2 and HeLa cells did not reveal any difference, suggesting that the change in stability in Caco-2 cells was not due to any eIF5A mutation. Pretreatment of cells with protease inhibitors such as phenylmethyl sulfonyl fluoride (PMSF) partially blocked the heat-induced loss of eIF5A and prevented heat-induced cell death. In light of the essential role of eIF5A in cell survival and proliferation, our results suggest that the stability of eIF5A may have an important role in determining the fate of the particular cell type after severe heat stress.


Subject(s)
Caco-2 Cells/physiology , Cell Death/physiology , Heat-Shock Response , Peptide Initiation Factors/metabolism , RNA-Binding Proteins/metabolism , Animals , Base Sequence , Caco-2 Cells/pathology , Cell Line , Cell Survival , DNA Fragmentation , Fibroblasts/cytology , Fibroblasts/physiology , Half-Life , Humans , Molecular Sequence Data , Oxidative Stress , Peptide Initiation Factors/genetics , Protease Inhibitors/metabolism , RNA-Binding Proteins/genetics , Sequence Alignment , Eukaryotic Translation Initiation Factor 5A
11.
J Cell Biochem ; 106(2): 267-78, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19097133

ABSTRACT

Induction of the heat shock response (HSR), determined by hsp70-luciferase reporter and HSP70 protein expression, is attenuated as a function of age of the IMR-90 human diploid fibroblasts. To better understand the underlying mechanism, we evaluated changes in the regulation and function of the HSF1 transcription factor. We show that the activation of HSF1 both in vivo and in vitro was decreased as a function of age, and this was attributable to a change in the regulation of HSF1 as the abundance of HSF1 protein and mRNA was unaffected. HSF1 was primarily cytosolic in young cells maintained at 37 degrees C, and heat shock promoted its quantitative nuclear translocation and trimerization. In old cells, some HSF1 was nuclear sequestered at 37 degrees C, and heat shock failed to promote the quantitative trimerization of HSF1. These changes in HSF1 could be reproduced by treating young cells with H2O2 to stunt them into premature senescence. Flow cytometry measurement of peroxide content showed higher levels in old cells and H2O2-induced premature senescent cells as compared to young cells. Experiments using isoelectric focusing and Western blot showed age-dependent changes in the mobility of HSF1 in a pattern consistent with its S-glutathiolation and S-nitrosylation; these changes could be mimicked by treating young cells with H2O2. Our results demonstrated dynamic age-dependent changes in the regulation but not the amount of HSF1. These changes are likely mediated by oxidative events that promote reversible and irreversible modification of HSF1 including S-glutathiolation and S-nitrosylation.


Subject(s)
Cellular Senescence/physiology , DNA-Binding Proteins/metabolism , Diploidy , Transcription Factors/metabolism , Cell Nucleus/metabolism , Cellular Senescence/drug effects , Fibroblasts , Genes, Reporter/genetics , Heat Shock Transcription Factors , Humans , Hydrogen Peroxide/pharmacology
12.
PLoS One ; 3(8): e2864, 2008 Aug 06.
Article in English | MEDLINE | ID: mdl-18682744

ABSTRACT

BACKGROUND: Induction of the heat shock response (HSR) and increased expression of the heat shock proteins (HSPs) provide mechanisms to ensure proper protein folding, trafficking, and disposition. The importance of HSPs is underscored by the understanding that protein mis-folding and aggregation contribute centrally to the pathogenesis of neurodegenerative diseases. METHODOLOGY/PRINCIPAL FINDINGS: We used a cell-based hsp70-luciferease reporter gene assay system to identify agents that modulate the HSR and show here that clinically relevant concentrations of the FDA-approved ALS drug riluzole significantly increased the heat shock induction of hsp70-luciferse reporter gene. Immuno-Western and -cytochemical analysis of HSF1 show that riluzole increased the amount of cytosolic HSF1 to afford a greater activation of HSF1 upon heat shock. The increased HSF1 contributed centrally to the cytoprotective activity of riluzole as hsf1 gene knockout negated the synergistic activity of riluzole and conditioning heat shock to confer cell survival under oxidative stress. Evidence of a post-transcriptional mechanism for the increase in HSF1 include: quantitation of mRNA(hsf1) by RT-PCR showed no effect of either heat shock or riluzole treatment; riluzole also increased the expression of HSF1 from a CMV-promoter; analysis of the turnover of HSF1 by pulse chase and immunoprecipitation show that riluzole slowed the decay of [(35)S]labeled-HSF1. The effect of riluzole on HSF1 was qualitatively different from that of MG132 and chloroquine, inhibitors of the proteasome and lysosome, respectively, and appeared to involve the chaperone-mediated autophagy pathway as RNAi-mediated knockdown of CMA negated its effect. CONCLUSION/SIGNIFICANCE: We show that riluzole increased the amount of HSF1 to amplify the HSR for cytoprotection. Our study provides novel insight into the mechanism that regulates HSF1 turnover, and identifies the degradation of HSF1 as a target for therapeutics intervention.


Subject(s)
DNA-Binding Proteins/genetics , Riluzole/pharmacology , Transcription Factors/genetics , Cell Survival/drug effects , DNA-Binding Proteins/biosynthesis , Excitatory Amino Acid Agonists/pharmacology , Genes, Reporter , HeLa Cells , Heat Shock Transcription Factors , Hot Temperature , Humans , Luciferases/genetics , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/physiopathology , Neurons/drug effects , Neurons/physiology , Spinal Cord/embryology , Spinal Cord/physiology , Transcription Factors/biosynthesis
13.
Cell Stress Chaperones ; 13(1): 73-84, 2008.
Article in English | MEDLINE | ID: mdl-18347944

ABSTRACT

Neuronal differentiation of the NG108-15 neuroblastoma-glioma hybrid cells is accompanied by a marked attenuation in the heat shock induction of the Hsp70-firefly luciferase reporter gene activity. Analysis of the amount and activation of heat shock factor 1, induction of mRNA(hsp), and the synthesis and accumulation of heat shock proteins (HSPs) in the undifferentiated and differentiated cells suggest a transcriptional mechanism for this attenuation. Concomitant with a decreased induction of the 72-kDa Hsp70 protein in the differentiated cells, there is an increased abundance of the constitutive 73-kDa Hsc70, a protein known to function in vesicle trafficking. Assessment of sensitivity of the undifferentiated and differentiated cells against stress-induced cell death reveals a significantly greater vulnerability of the differentiated cells toward the cytotoxic effects of arsenite and glutamate/glycine. This study shows that changes in regulation of the HSP and HSC proteins are components of the neuronal cell differentiation program and that the attenuated induction of HSPs likely contributes to neuronal vulnerability whereas the increased expression of Hsc70 likely has a role in neural-specific functions.


Subject(s)
Heat-Shock Proteins/genetics , Neurons/metabolism , Animals , Bucladesine/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Genes, Reporter , Glioma/pathology , HSP70 Heat-Shock Proteins/biosynthesis , HSP70 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/biosynthesis , HSP90 Heat-Shock Proteins/genetics , Heat-Shock Proteins/biosynthesis , Hippocampus/cytology , Hippocampus/embryology , Hot Temperature , Hybrid Cells/cytology , Hybrid Cells/drug effects , Hybrid Cells/metabolism , Mice , Molecular Chaperones , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neurites/ultrastructure , Neuroblastoma/pathology , Neurons/cytology , Neurons/drug effects , RNA, Messenger/biosynthesis , Rats , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Transcription, Genetic
14.
Brain Res ; 1203: 39-50, 2008 Apr 08.
Article in English | MEDLINE | ID: mdl-18316066

ABSTRACT

Differentiation of neural progenitor cells of neuroblastoma, pheochromocytoma, and surrogate stem cell lineages from a state resembling stem cells to a state resembling neurons is accompanied by a marked attenuation in induction of the heat shock protein 70 promoter driven-luciferase reporter gene, and induction of the reporter gene in primary embryonic neurons from hippocampus, cortex, and spinal cord is lower still when compared to the differentiated cells. Neural specificity of this phenotype is demonstrated by a negative correlation of hsp70-reporter gene expression and neurite extension under various experimental conditions. Analysis of biochemical events involved in induction of the heat shock response (HSR) reveal a blunted activation of HSF1 DNA-binding activity, and decreased induction of the mRNA(hsp70) and the 72 kDa HSP70 protein. Immunocytochemical staining for HSP70 demonstrates a cytoplasmic staining pattern; heat shock greatly increased the HSP70 staining intensity in the undifferentiated cells and less so in the differentiated cells. Vulnerability of the differentiated cells towards the oxidizer, arsenite, and the excitotoxic glutamate/glycine is demonstrated by the dose-dependent cytotoxic effects of these agents on cell viability and activation of caspase 3/7. Importantly, conditioning heat shock as well as increased expression of HSP70 by gene transfer conferred protection against such cytotoxicity. Together, our results show that neural differentiation is associated with a decreased induction of the heat shock response and an increased vulnerability to stress induced pathologies and death.


Subject(s)
Cell Differentiation/physiology , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Response/physiology , Neurons/physiology , Animals , Arsenites/pharmacology , Axons/physiology , Brain/cytology , Bucladesine/pharmacology , Cell Differentiation/drug effects , Cell Survival , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Electric Stimulation/methods , Embryo, Mammalian , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Gene Expression Regulation/radiation effects , HSP70 Heat-Shock Proteins/genetics , Heat Shock Transcription Factors , Heat-Shock Response/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Mice , Nerve Tissue Proteins/metabolism , Neuroblastoma , Neurons/cytology , Neurons/drug effects , Patch-Clamp Techniques , Rats , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism
15.
Clin Dermatol ; 24(5): 393-413, 2006.
Article in English | MEDLINE | ID: mdl-16966020

ABSTRACT

Sjogren's Syndrome (SS) is a systemic autoimmune disease characterized by dry eyes (keratoconjunctivis sicca) and dry mouth (xerostomia). To fulfill diagnostic criteria, patients must have objective signs of dryness on examination and laboratory confirmation of an autoimmune process as evidenced by a positive autoantibody to SS-A antigen or a characteristic lip biopsy. SS may exist as a primary condition or in association with other systemic autoimmune disorders (termed secondary SS) such as rheumatoid arthritis, systemic lupus erythematous (SLE), progressive systemic sclerosis (scleroderma), or dermatomyositis. Exclusions to the diagnosis include pre-existing lymphoma, hepatitis C or HIV infection. Pathogenesis involves both genetic (especially HLA-DR) and environmental factors. Both T-cells and B-cells are involved in the generation of cytokines and chemokines within the glands. The epithelial cells of the glands also play a role in pathogenesis. The dermatologic manifestations range from drynessness (sicca) and its complications to vasculitis. There is a significant overlap in the clinical manifestations, as well as treatment, of SS and SLE. However, SS patients require special attention to the complications of ocular dryness (keratocojunctivitis sicca and blepharitis) and oral dryness (rapid tooth loss and oral candidiasis) SS patients have a markedly increased risk of lymphoma and enlarged lymph nodes or persistently enlarged parotid/submandibular glands that require further evaluation.


Subject(s)
Autoimmune Diseases/pathology , Sjogren's Syndrome/pathology , Skin Diseases/pathology , Autoimmune Diseases/diagnosis , Autoimmune Diseases/therapy , Diagnosis, Differential , Humans , Immunosuppressive Agents/therapeutic use , Keratoconjunctivitis Sicca/etiology , Keratoconjunctivitis Sicca/pathology , Keratoconjunctivitis Sicca/physiopathology , Keratoconjunctivitis Sicca/therapy , Lubrication , Lupus Erythematosus, Systemic/diagnosis , Sjogren's Syndrome/diagnosis , Sjogren's Syndrome/therapy , Skin/immunology , Skin/pathology , Skin/physiopathology , Skin Diseases/diagnosis , Skin Diseases/therapy , Vasculitis/drug therapy , Vasculitis/etiology , Vasculitis/pathology , Xerostomia/etiology , Xerostomia/pathology , Xerostomia/physiopathology , Xerostomia/therapy
16.
J Cell Physiol ; 207(2): 562-9, 2006 May.
Article in English | MEDLINE | ID: mdl-16447264

ABSTRACT

The objective of this study is to better define induction of the heat shock response by arsenite, and to evaluation if induction of heat shock proteins (HSPs) contributes to the carcinogenic activity of arsenite. We show here that arsenite is a ubiquitous inducer of the heat shock response in mammalian cells: that it activated heat shock transcription factor 1 (HSF1) DNA-binding activity, enhanced hsp 70 promoter, and induced hsp70mRNA and synthesis of HSP chaperones. Using a high throughput hsp70 promoter-luciferase reporter assay, we observed a hormetic dose response where low concentrations of arsenite stimulated and high concentrations inhibited. Further, the response was time-dependent such that with longer times of incubation, the dose response shifted to the left. The effect of arsenite in inducing the hsp 70-luciferase reporter absolutely required a functional HSF1 as it was not observed in HSF1 minus cells but re-instated by expression of HSF1. Consistent with the suggestion that arsenic targets vicinal cysteine-SH, we showed that dithiothreitol blocked the effect of arsenite. Assays of cell viability and caspase showed that arsenite caused a dose-dependent increase in cell death by activation of caspase 3/7 and pre-induction of HSPs blunted these effects. Using anchorage independent cell growth as a late stage tumor promotion assay, we showed that low concentrations of arsenite had a growth promoting effect, which was enhanced by moderate heat shock. Our study provides evidence that induction of the heat shock response is a sensitive biomarker of arsenic exposure and that induction of HSPs likely contributes to the tumor promotion effect of arsenic.


Subject(s)
Arsenites/toxicity , Heat-Shock Response/drug effects , Neoplasms/chemically induced , Animals , Caspase 3 , Caspase 7 , Caspases/metabolism , Cell Line , Cell Line, Transformed , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dithiothreitol/pharmacology , Dose-Response Relationship, Drug , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression/drug effects , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , HeLa Cells , Heat Shock Transcription Factors , Humans , Mice , Mice, Knockout , NIH 3T3 Cells , Neoplasms/genetics , Neoplasms/metabolism , Sulfhydryl Compounds/pharmacology , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/metabolism
17.
Article in Chinese | MEDLINE | ID: mdl-12796815

ABSTRACT

To evaluate the effects of cysteine-SH-directed regents on the redox status, structure and function of human heat shock transcription factor 1 (hHSF1), treatment in vitro of hHSF1 with 0.3 0.5 mmol/L oxidizing reagent diamide (DM) and treatment in vivo of HeLa cells with 1 mmol/L buthionine sulfoximine (BSO), an inhibitor of gamma-glutamylcysteine synthetase, promoted the formation of a compact, intramolecularly disulfide-crosslinked, stable monomeric form of ox-hHSF1, and blocked the trimerization and activation of HSF1. The effects of diamine were dose-dependent and readily could be completely reversed by adding 0.4 0.5 mmol/L reducing reagent dithiothreitol (DTT) to the samples prior to gel electrophoresis. Computer modeling of the alpha-helical coiled-coil domains of the HSF1 monomer and trimer showed that the alignment of the N- and C-terminal hydrophobic repeats of HSF1 monomer could bring C(3)(Cys(153))close to C(4) and C(5)(Cys(373) and Cys(378), respectively), in positions permissible for disulfide bond formation under appropriate experimental conditions. The results suggest that redox-dependent thiol-disulfide exchange can provide a mechanism for regulation the conformation and activity of hHSF1.


Subject(s)
DNA-Binding Proteins/metabolism , Heat-Shock Proteins/metabolism , Buthionine Sulfoximine/pharmacology , DNA-Binding Proteins/chemistry , Diamide/pharmacology , Dithiothreitol/pharmacology , Dose-Response Relationship, Drug , Glutamate-Cysteine Ligase/antagonists & inhibitors , HeLa Cells , Heat Shock Transcription Factors , Heat-Shock Proteins/chemistry , Humans , Models, Molecular , Oxidation-Reduction , Protein Binding/drug effects , Protein Conformation , Transcription Factors
18.
J Invest Dermatol ; 118(2): 275-81, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11841544

ABSTRACT

Intact human epidermis resists invasion by pathogenic microbes but the biochemical basis of its resistance is not well understood. Recently, an antimicrobial peptide, human beta-defensin-2, was discovered in inflamed epidermis. We used a recombinant baculovirus/insect cell system to produce human beta-defensin-2 and confirmed that at micromolar concentrations it has a broad spectrum of antimicrobial activity, with the striking exception of Staphylococcus aureus. Immunostaining with a polyclonal antibody to human beta-defensin-2 showed that the expression of human beta-defensin-2 peptide by human keratinocytes required differentiation of the cells (either by increased calcium concentration or by growth and maturation in epidermal organotypic culture) as well as a cytokine or bacterial stimulus. Interleukin-1alpha, interleukin-1beta, or live Pseudomonas aeruginosa proved to be the most effective stimuli whereas other bacteria and cytokines had little or no ability to induce human beta-defensin-2 synthesis. In interleukin-1alpha-stimulated epidermal cultures, human beta-defensin-2 first appeared in the cytoplasm in differentiated suprabasal layers of skin, next in a more peripheral web-like distribution in the upper layers of the epidermis, and then over a few days migrated to the stratum corneum. By semiquantitative Western blot analysis of epidermal lysates, the average concentration of human beta-defensin-2 in stimulated organotypic epidermal culture reached 15--70 microg per gram of tissue, i.e., 3.5-16 microM, well within the range required for antimicrobial activity. Because of the restricted pattern of human beta-defensin-2 distribution in the epidermis, its local concentration must be much higher. Defensins and other antimicrobial peptides of inflamed epidermis are likely to play an important antimicrobial role in host defense against cutaneous pathogens.


Subject(s)
Bacterial Physiological Phenomena , Interleukin-1/physiology , Keratinocytes/metabolism , Keratinocytes/microbiology , beta-Defensins/biosynthesis , Anti-Bacterial Agents/metabolism , Cell Differentiation/physiology , Cells, Cultured , Humans , Keratinocytes/cytology , Organ Culture Techniques , Recombinant Proteins/metabolism , Tissue Distribution , beta-Defensins/metabolism
19.
Biochemistry ; 41(8): 2580-8, 2002 Feb 26.
Article in English | MEDLINE | ID: mdl-11851405

ABSTRACT

We present here evidence that redox-dependent thiol-disulfide exchange can provide a mechanism regulating the conformation and activity of the human heat shock transcription factor 1 (HSF1). Diamide and dithiothreitol were reagents used respectively to promote and reverse disulfide cross-link, and the resolution and detection of redox conformers of HSF1 were done according to recently published methods [Manalo, D. J., and Liu, A. Y.-C. (2001) J. Biol. Chem. 276, 23554-23561]. We showed that preincubation of the latent HSF1 monomer with diamide inhibited the in vitro heat-induced activation and trimerization of HSF1 and caused the formation of ox-hHSF1, a compact, disulfide cross-linked HSF1 conformer detectable in immuno-Western blot assay. These effects of diamide were dose-dependent and were rapidly and quantitatively reversed by dithiothreitol. Cysteine site-specific mutants of HSF1 were constructed and used to determine which of the five cysteine residues may be engaged in disulfide cross-link. Analysis of the in vitro transcribed and translated HSF1 proteins showed that while mutation of C1 (amino acid 36) and C2 (amino acid 103) had no effect on the redox sensitivity of HSF1, the mutation of C3 (amino acid 153) or double mutation of C4 and C5 (amino acids 373 and 378, respectively) to serine rendered HSF1 insensitive to diamide and prevented its conversion to ox-HSF1. HSF1 with a single cysteine to serine mutation at either the C4 or C5 position gave different ox-HSF1 conformers in the presence of diamide, suggesting that C3 could be disulfide cross-linked to either C4 or C5. The possibility that HSF1 may have integrated redox chemistry of cysteine sulfhydryl into its functional response in higher mammalian cells is discussed.


Subject(s)
DNA-Binding Proteins/metabolism , Base Sequence , Cysteine/chemistry , Cysteine/metabolism , DNA Primers , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Disulfides/chemistry , Electrophoresis, Polyacrylamide Gel , HeLa Cells , Heat Shock Transcription Factors , Humans , Models, Molecular , Mutagenesis, Site-Directed , Oxidation-Reduction , Protein Conformation , Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...