Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
2.
Cell Stress Chaperones ; 28(1): 1-9, 2023 01.
Article in English | MEDLINE | ID: mdl-36602710

ABSTRACT

The Second International Symposium on Cellular and Organismal Stress Responses took place virtually on September 8-9, 2022. This meeting was supported by the Cell Stress Society International (CSSI) and organized by Patricija Van Oosten-Hawle and Andrew Truman (University of North Carolina at Charlotte, USA) and Mehdi Mollapour (SUNY Upstate Medical University, USA). The goal of this symposium was to continue the theme from the initial meeting in 2020 by providing a platform for established researchers, new investigators, postdoctoral fellows, and students to present and exchange ideas on various topics on cellular stress and chaperones. We will summarize the highlights of the meeting here and recognize those that received recognition from the CSSI.


Subject(s)
Molecular Chaperones , Stress, Physiological , Humans , HSP70 Heat-Shock Proteins , Molecular Chaperones/physiology , Stress, Physiological/physiology
3.
Cell Death Dis ; 13(12): 1038, 2022 12 13.
Article in English | MEDLINE | ID: mdl-36513635

ABSTRACT

Prenatal inflammatory insults accompany prematurity and provoke diffuse white matter injury (DWMI), which is associated with increased risk of neurodevelopmental pathologies, including autism spectrum disorders. DWMI results from maturation arrest of oligodendrocyte precursor cells (OPCs), a process that is poorly understood. Here, by using a validated mouse model of OPC maturation blockade, we provide the genome-wide ID card of the effects of neuroinflammation on OPCs that reveals the architecture of global cell fate issues underlining their maturation blockade. First, we find that, in OPCs, neuroinflammation takes advantage of a primed epigenomic landscape and induces abnormal overexpression of genes of the immune/inflammatory pathways: these genes strikingly exhibit accessible chromatin conformation in uninflamed OPCs, which correlates with their developmental, stage-dependent expression, along their normal maturation trajectory, as well as their abnormal upregulation upon neuroinflammation. Consistently, we observe the positioning on DNA of key transcription factors of the immune/inflammatory pathways (IRFs, NFkB), in both unstressed and inflamed OPCs. Second, we show that, in addition to the general perturbation of the myelination program, neuroinflammation counteracts the physiological downregulation of the cell cycle pathway in maturing OPCs. Neuroinflammation therefore perturbs cell identity in maturing OPCs, in a global manner. Moreover, based on our unraveling of the activity of genes of the immune/inflammatory pathways in prenatal uninflamed OPCs, the mere suppression of these proinflammatory mediators, as currently proposed in the field, may not be considered as a valid neurotherapeutic strategy.


Subject(s)
Oligodendroglia , White Matter , Mice , Animals , Pregnancy , Female , Oligodendroglia/metabolism , Mice, Transgenic , White Matter/pathology , Epigenomics , Mice, Inbred C57BL , Neuroinflammatory Diseases , Cell Differentiation , Cell Cycle/genetics , Epigenesis, Genetic
4.
Nat Commun ; 13(1): 7002, 2022 11 16.
Article in English | MEDLINE | ID: mdl-36385105

ABSTRACT

Patients carrying autosomal dominant mutations in the histone/lysine acetyl transferases CBP or EP300 develop a neurodevelopmental disorder: Rubinstein-Taybi syndrome (RSTS). The biological pathways underlying these neurodevelopmental defects remain elusive. Here, we unravel the contribution of a stress-responsive pathway to RSTS. We characterize the structural and functional interaction between CBP/EP300 and heat-shock factor 2 (HSF2), a tuner of brain cortical development and major player in prenatal stress responses in the neocortex: CBP/EP300 acetylates HSF2, leading to the stabilization of the HSF2 protein. Consequently, RSTS patient-derived primary cells show decreased levels of HSF2 and HSF2-dependent alteration in their repertoire of molecular chaperones and stress response. Moreover, we unravel a CBP/EP300-HSF2-N-cadherin cascade that is also active in neurodevelopmental contexts, and show that its deregulation disturbs neuroepithelial integrity in 2D and 3D organoid models of cerebral development, generated from RSTS patient-derived iPSC cells, providing a molecular reading key for this complex pathology.


Subject(s)
CREB-Binding Protein , Heat-Shock Proteins , Neurodevelopmental Disorders , Rubinstein-Taybi Syndrome , Transcription Factors , Humans , CREB-Binding Protein/genetics , CREB-Binding Protein/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Histones/genetics , Mutation , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/pathology , Rubinstein-Taybi Syndrome/genetics , Rubinstein-Taybi Syndrome/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , E1A-Associated p300 Protein/genetics , E1A-Associated p300 Protein/metabolism
5.
Cell Stress Chaperones ; 26(5): 819-833, 2021 09.
Article in English | MEDLINE | ID: mdl-34331200

ABSTRACT

Cancer cells rely on heat shock proteins (HSPs) for growth and survival. Especially HSP90 has multiple client proteins and plays a critical role in malignant transformation, and therefore different types of HSP90 inhibitors are being developed. The bioactive natural compound gambogic acid (GB) is a prenylated xanthone with antitumor activity, and it has been proposed to function as an HSP90 inhibitor. However, there are contradicting reports whether GB induces a heat shock response (HSR), which is cytoprotective for cancer cells and therefore a potentially problematic feature for an anticancer drug. In this study, we show that GB and a structurally related compound, called gambogenic acid (GBA), induce a robust HSR, in a thiol-dependent manner. Using heat shock factor 1 (HSF1) or HSF2 knockout cells, we show that the GB or GBA-induced HSR is HSF1-dependent. Intriguingly, using closed form ATP-bound HSP90 mutants that can be co-precipitated with HSF1, a known facilitator of cancer, we show that also endogenous HSF2 co-precipitates with HSP90. GB and GBA treatment disrupt the interaction between HSP90 and HSF1 and HSP90 and HSF2. Our study implies that these compounds should be used cautiously if developed for cancer therapies, since GB and its derivative GBA are strong inducers of the HSR, in multiple cell types, by involving the dissociation of a HSP90-HSF1/HSF2 complex.


Subject(s)
Heat Shock Transcription Factors/metabolism , Heat-Shock Proteins/metabolism , Heat-Shock Response , Sulfhydryl Compounds/metabolism , Transcription Factors/metabolism , Xanthenes/pharmacology , Xanthones/pharmacology , CRISPR-Cas Systems/genetics , Cell Line, Tumor , HSP90 Heat-Shock Proteins/metabolism , Heat-Shock Response/drug effects , Humans , Protein Binding/drug effects , Xanthenes/chemistry , Xanthones/chemistry
6.
Addict Biol ; 26(2): e12939, 2021 03.
Article in English | MEDLINE | ID: mdl-32720424

ABSTRACT

Ethanol consumption impairs learning and memory through disturbances of NMDA-type glutamate receptor-dependent synaptic plasticity (long-term depression [LTD] and long-term potentiation [LTP]) in the hippocampus. Recently, we demonstrated that two ethanol binge-like episodes in young adult rats selectively blocked NMDA-LTD in hippocampal slices, increased NMDA receptor sensitivity to a GluN2B subunit antagonist, and induced cognitive deficits. Here, using knockout adult mice, we show that a stress-responsive transcription factor of the heat shock factor family, HSF2, which is involved in the perturbation of brain development induced by ethanol, participates in these processes. In the absence of ethanol, hsf2-/- mice show a selective loss of LTD in the hippocampus, which is associated with an increased sensitivity of NMDA-field excitatory postsynaptic potentials (fEPSPs) to a GluN2B antagonist, compared with wild-type (WT) mice. These results suggest that HSF2 is required for proper glutamatergic synaptic transmission and LTD plasticity. After 1 month of chronic ethanol consumption in a two-bottle choice paradigm, WT mice showed an increase in hippocampal synaptic transmission, an enhanced sensitivity to GluN2B antagonist, and a blockade of LTD. In contrast, such modulation of synaptic transmission and plasticity were absent in hsf2-/- mice. We conclude that HSF2 is an important mediator of both glutamatergic neurotransmission and synaptic plasticity in basal conditions and also mediates ethanol-induced neuroadaptations of the hippocampus network after chronic ethanol intake.


Subject(s)
Ethanol/pharmacology , Heat Shock Transcription Factors/drug effects , Long-Term Potentiation/drug effects , Long-Term Synaptic Depression/drug effects , N-Methylaspartate/drug effects , Adolescent , Adult , Age Factors , Animals , Hippocampus/drug effects , Humans , Mice
7.
Neurosci Lett ; 725: 134895, 2020 04 23.
Article in English | MEDLINE | ID: mdl-32147500

ABSTRACT

The Heat Shock Factors (HSFs) have been historically identified as a family of transcription factors that are activated and work in a stress-responsive manner, after exposure to a large variety of stimuli. However, they are also critical in normal conditions, in a life long manner, in a number of physiological processes that encompass gametogenesis, embryonic development and the integrity of adult organs and organisms. The importance of such roles is emphasized by the devastating impact of their deregulation on health, ranging from reproductive failure, neurodevelopmental disorders, cancer, and aging pathologies, including neurodegenerative disorders. Here, we provide an overview of the delicate choreography of the regulation of HSFs during neurodevelopment, at prenatal and postnatal stages. The regulation of HSFs acts at multiple layers and steps, and comprises the control of (i) HSF mRNA and protein levels, (ii) HSF activity in terms of DNA-binding and transcription, (iii) HSF homo- and hetero-oligomerization capacities, and (iv) HSF combinatory set of post-translational modifications. We also describe how these regulatory mechanisms operate in the normal developing brain and how their perturbation impact neurodevelopment under prenatal or perinatal stress conditions. In addition, we put into perspective the possible role of HSFs in the evolution of the vertebrate brains and the importance of the HSF pathway in a large variety of neurodevelopmental disorders.


Subject(s)
Brain/growth & development , Brain/metabolism , Heat Shock Transcription Factors/metabolism , Heat-Shock Proteins/metabolism , Animals , Brain/physiopathology , Heat-Shock Proteins/genetics , Heat-Shock Response/physiology , Humans , Transcription, Genetic/physiology
8.
Cell Rep ; 30(2): 583-597.e6, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31940498

ABSTRACT

Maintenance of protein homeostasis, through inducible expression of molecular chaperones, is essential for cell survival under protein-damaging conditions. The expression and DNA-binding activity of heat shock factor 2 (HSF2), a member of the heat shock transcription factor family, increase upon exposure to prolonged proteotoxicity. Nevertheless, the specific roles of HSF2 and the global HSF2-dependent gene expression profile during sustained stress have remained unknown. Here, we found that HSF2 is critical for cell survival during prolonged proteotoxicity. Strikingly, our RNA sequencing (RNA-seq) analyses revealed that impaired viability of HSF2-deficient cells is not caused by inadequate induction of molecular chaperones but is due to marked downregulation of cadherin superfamily genes. We demonstrate that HSF2-dependent maintenance of cadherin-mediated cell-cell adhesion is required for protection against stress induced by proteasome inhibition. This study identifies HSF2 as a key regulator of cadherin superfamily genes and defines cell-cell adhesion as a determinant of proteotoxic stress resistance.


Subject(s)
Cell Death/immunology , Cell Survival/immunology , Heat Shock Transcription Factors/metabolism , Heat-Shock Proteins/metabolism , Animals , Cell Adhesion , Humans , Up-Regulation
9.
Pediatr Res ; 85(2): 155-165, 2019 01.
Article in English | MEDLINE | ID: mdl-30446768

ABSTRACT

Genetic anomalies have a role in autism spectrum disorders (ASD). Each genetic factor is responsible for a small fraction of cases. Environment factors, like preterm delivery, have an important role in ASD. Preterm infants have a 10-fold higher risk of developing ASD. Preterm birth is often associated with maternal/fetal inflammation, leading to a fetal/neonatal inflammatory syndrome. There are demonstrated experimental links between fetal inflammation and the later development of behavioral symptoms consistent with ASD. Preterm infants have deficits in connectivity. Most ASD genes encode synaptic proteins, suggesting that ASD are connectivity pathologies. Microglia are essential for normal synaptogenesis. Microglia are diverted from homeostatic functions towards inflammatory phenotypes during perinatal inflammation, impairing synaptogenesis. Preterm infants with ASD have a different phenotype from term born peers. Our original hypothesis is that exposure to inflammation in preterm infants, combined with at risk genetic background, deregulates brain development leading to ASD.


Subject(s)
Autism Spectrum Disorder/physiopathology , Central Nervous System Diseases/physiopathology , Infant, Premature , Inflammation/physiopathology , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Central Nervous System Diseases/genetics , Central Nervous System Diseases/pathology , Humans , Infant, Newborn , Inflammation/pathology
10.
Nat Commun ; 9(1): 1431, 2018 04 12.
Article in English | MEDLINE | ID: mdl-29650953

ABSTRACT

Heat shock protein 27 (HSP27/HSPB1) is a stress-inducible chaperone that facilitates cancer development by its proliferative and anti-apoptotic functions. The OGX-427 antisense oligonucleotide against HSP27 has been reported to be beneficial against idiopathic pulmonary fibrosis. Here we show that OGX-427 is effective in two murine models of thrombopoietin- and JAKV617F-induced myelofibrosis. OGX-427 limits disease progression and is associated with a reduction in spleen weight, in megakaryocyte expansion and, for the JAKV617F model, in fibrosis. HSP27 regulates the proliferation of JAK2V617F-positive cells and interacts directly with JAK2/STAT5. We also show that its expression is increased in both CD34+ circulating progenitors and in the serum of patients with JAK2-dependent myeloproliferative neoplasms with fibrosis. Our data suggest that HSP27 plays a key role in the pathophysiology of myelofibrosis and represents a new potential therapeutic target for patients with myeloproliferative neoplasms.


Subject(s)
HSP27 Heat-Shock Proteins/genetics , Janus Kinase 2/genetics , Oligonucleotides/pharmacology , Primary Myelofibrosis/drug therapy , Primary Myelofibrosis/genetics , STAT5 Transcription Factor/genetics , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Bone Marrow Transplantation , Cell Line, Tumor , Disease Models, Animal , Female , HEK293 Cells , HSP27 Heat-Shock Proteins/immunology , Humans , Janus Kinase 2/immunology , K562 Cells , Leukocytes/drug effects , Leukocytes/immunology , Leukocytes/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Targeted Therapy , Mutation , Primary Myelofibrosis/immunology , Primary Myelofibrosis/pathology , STAT5 Transcription Factor/immunology , Thrombopoietin/genetics , Thrombopoietin/immunology , Transduction, Genetic , Whole-Body Irradiation
11.
Cell Stress Chaperones ; 23(2): 171-177, 2018 03.
Article in English | MEDLINE | ID: mdl-29396663

ABSTRACT

About 150 international scientists gathered in Turku, Finland, in August of 2017 for the eighth in a series of international congresses about the roles of stress proteins in biology and medicine. The scientific theme and title of the 2017 Congress was "Stress Management Mechanisms and Pathways." The meeting covered a broad range of topics, reflecting the wide scope of the Cell Stress Society International (CSSI) and highlighting the numerous recent breakthroughs in stress response biology and medicine. The keynote lecturers included Marja Jäättelä, Richard Morimoto, Anne Bertolotti, and Peter Walter. The Executive Council of the CSSI elected new Fellows and Senior Fellows. The Spirit of Budapest Award was presented to Peter Csermely, Wolfgang Schumann, and Subhash Lakhotia in recognition of pioneering service contributions to the CSSI. The CSSI Medallion for Career Achievement was awarded to Larry Hightower and CSSI president Gabriella Santoro proclaimed Tuesday, August 15, 2017, Robert M. Tanguay Day at the congress in recognition of Robert's many years of scientific accomplishment and work on behalf of the CSSI. Additional special events were the awarding of the Ferruccio Ritossa Early Career Award to Serena Carra and the Alfred Tissières Young Investigator Award to Ayesha Murshid. As is the tradition at CSSI congresses, there were social events that included an exciting piano performance by a trio of young Finnish pianists, at the Sibelius Museum.


Subject(s)
Biology , Medicine , Animals , Caenorhabditis elegans/physiology , Gene Regulatory Networks , Heat-Shock Proteins , Humans , Longevity , Oxidative Stress , Protein Aggregates , Proteostasis , Stress, Physiological
12.
Cell Stress Chaperones ; 23(1): 115-126, 2018 01.
Article in English | MEDLINE | ID: mdl-28712054

ABSTRACT

Abundant evidence has accumulated showing that fetal alcohol exposure broadly modifies DNA methylation profiles in the brain. DNA methyltransferases (DNMTs), the enzymes responsible for DNA methylation, are likely implicated in this process. However, their regulation by ethanol exposure has been poorly addressed. Here, we show that alcohol exposure modulates DNMT protein levels through multiple mechanisms. Using a neural precursor cell line and primary mouse embryonic fibroblasts (MEFs), we found that ethanol exposure augments the levels of Dnmt3a, Dnmt3b, and Dnmt3l transcripts. We also unveil similar elevation of mRNA levels for other epigenetic actors upon ethanol exposure, among which the induction of lysine demethylase Kdm6a shows heat shock factor dependency. Furthermore, we show that ethanol exposure leads to specific increase in DNMT3A protein levels. This elevation not only relies on the upregulation of Dnmt3a mRNA but also depends on posttranscriptional mechanisms that are mediated by NADPH oxidase-dependent production of reactive oxygen species (ROS). Altogether, our work underlines complex regulation of epigenetic actors in response to alcohol exposure at both transcriptional and posttranscriptional levels. Notably, the upregulation of DNMT3A emerges as a prominent molecular event triggered by ethanol, driven by the generation of ROS.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , Ethanol/adverse effects , Reactive Oxygen Species/metabolism , Up-Regulation , Animals , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Heat Shock Transcription Factors/metabolism , Mice , NADPH Oxidases/metabolism , Neural Stem Cells/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
13.
Nat Commun ; 8: 15157, 2017 05 02.
Article in English | MEDLINE | ID: mdl-28462912

ABSTRACT

Repetitive prenatal exposure to identical or similar doses of harmful agents results in highly variable and unpredictable negative effects on fetal brain development ranging in severity from high to little or none. However, the molecular and cellular basis of this variability is not well understood. This study reports that exposure of mouse and human embryonic brain tissues to equal doses of harmful chemicals, such as ethanol, activates the primary stress response transcription factor heat shock factor 1 (Hsf1) in a highly variable and stochastic manner. While Hsf1 is essential for protecting the embryonic brain from environmental stress, excessive activation impairs critical developmental events such as neuronal migration. Our results suggest that mosaic activation of Hsf1 within the embryonic brain in response to prenatal environmental stress exposure may contribute to the resulting generation of phenotypic variations observed in complex congenital brain disorders.


Subject(s)
Brain/drug effects , Heat Shock Transcription Factors/genetics , Neural Stem Cells/drug effects , Neurons/drug effects , Prenatal Exposure Delayed Effects/genetics , Adult , Animals , Brain/growth & development , Brain/metabolism , Brain/pathology , Cell Movement/drug effects , Embryo, Mammalian , Ethanol/pharmacology , Female , Gene Expression Regulation, Developmental , Heat Shock Transcription Factors/metabolism , Humans , Hydrogen Peroxide/pharmacology , Injections, Intraperitoneal , Male , Maternal Exposure/adverse effects , Mice , Mice, Transgenic , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Phenotype , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/pathology , Primary Cell Culture , Signal Transduction
14.
J Mol Biol ; 427(24): 3793-816, 2015 Dec 04.
Article in English | MEDLINE | ID: mdl-26482101

ABSTRACT

Starting as a paradigm for stress responses, the study of the transcription factor (TF) family of heat shock factors (HSFs) has quickly and widely expanded these last decades, thanks to their fascinating and significant involvement in a variety of pathophysiological processes, including development, reproduction, neurodegeneration and carcinogenesis. HSFs, originally defined as classical TFs, strikingly appeared to play a central and often pioneering role in reshaping the epigenetic landscape. In this review, we describe how HSFs are able to sense the epigenetic environment, and we review recent data that support their role as sculptors of the chromatin landscape through their complex interplay with chromatin remodelers, histone-modifying enzymes and non-coding RNAs.


Subject(s)
Epigenesis, Genetic , Heat-Shock Proteins/physiology , Stress, Physiological , Transcription Factors/physiology , Animals , Base Sequence , Binding Sites , Humans , Transcription, Genetic
15.
Cell Stress Chaperones ; 19(6): 753-61, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25199949

ABSTRACT

A collaborative workshop dedicated to the discussion of heat shock factors in stress response, development, and disease was held on April 22-24, 2014 at the Université Paris Diderot in Paris, France. Recent years have witnessed an explosion of interest in these highly conserved transcription factors, with biological roles ranging from environmental sensing to human development and cancer.


Subject(s)
Biomedical Research , Heat Stress Disorders/metabolism , Heat-Shock Proteins/metabolism , Heat-Shock Response , Transcription Factors/metabolism , Animals , Gene Expression Regulation , Heat Stress Disorders/genetics , Heat Stress Disorders/physiopathology , Heat-Shock Proteins/genetics , Humans , Signal Transduction , Transcription Factors/genetics
16.
EMBO Mol Med ; 6(8): 1043-61, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25027850

ABSTRACT

Fetal alcohol spectrum disorder (FASD) is a frequent cause of mental retardation. However, the molecular mechanisms underlying brain development defects induced by maternal alcohol consumption during pregnancy are unclear. We used normal and Hsf2-deficient mice and cell systems to uncover a pivotal role for heat shock factor 2 (HSF2) in radial neuronal migration defects in the cortex, a hallmark of fetal alcohol exposure. Upon fetal alcohol exposure, HSF2 is essential for the triggering of HSF1 activation, which is accompanied by distinctive post-translational modifications, and HSF2 steers the formation of atypical alcohol-specific HSF1-HSF2 heterocomplexes. This perturbs the in vivo binding of HSF2 to heat shock elements (HSEs) in genes that control neuronal migration in normal conditions, such as p35 or the MAPs (microtubule-associated proteins, such as Dclk1 and Dcx), and alters their expression. In the absence of HSF2, migration defects as well as alterations in gene expression are reduced. Thus, HSF2, as a sensor for alcohol stress in the fetal brain, acts as a mediator of the neuronal migration defects associated with FASD.


Subject(s)
Fetal Alcohol Spectrum Disorders/pathology , Heat-Shock Proteins/metabolism , Malformations of Cortical Development, Group II/chemically induced , Stress, Physiological , Transcription Factors/metabolism , Animals , Cerebral Cortex/pathology , DNA-Binding Proteins/metabolism , Disease Models, Animal , Doublecortin Protein , Gene Expression Regulation , Heat Shock Transcription Factors , Heat-Shock Proteins/deficiency , Mice , Mice, Knockout , Protein Binding , Transcription Factors/deficiency
17.
PLoS Biol ; 12(6): e1001890, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24960041

ABSTRACT

During early development, modulations in the expression of Nodal, a TGFß family member, determine the specification of embryonic and extra-embryonic cell identities. Nodal has been extensively studied in the mouse, but aspects of its early expression remain unaccounted for. We identified a conserved hotspot for the binding of pluripotency factors at the Nodal locus and called this sequence "highly bound element" (HBE). Luciferase-based assays, the analysis of fluorescent HBE reporter transgenes, and a conditional mutation of HBE allowed us to establish that HBE behaves as an enhancer, is activated ahead of other Nodal enhancers in the epiblast, and is essential to Nodal expression in embryonic stem cells (ESCs) and in the mouse embryo. We also showed that HBE enhancer activity is critically dependent on its interaction with the pluripotency factor Oct4 and on Activin/Nodal signaling. Use of an in vitro model of epiblast maturation, relying on the differentiation of ESCs into epiblast stem cells (EpiSCs), revealed that this process entails a shift in the regulation of Nodal expression from an HBE-driven phase to an ASE-driven phase, ASE being another autoregulatory Nodal enhancer. Deletion of HBE in ESCs or in EpiSCs allowed us to show that HBE, although not necessary for Nodal expression in EpiSCs, is required in differentiating ESCs to activate the differentiation-promoting ASE and therefore controls this regulatory shift. Our findings clarify how early Nodal expression is regulated and suggest how this regulation can promote the specification of extra-embryonic precusors without inducing premature differentiation of epiblast cells. More generally, they open new perspectives on how pluripotency factors achieve their function.


Subject(s)
Embryonic Stem Cells/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Germ Layers/physiology , Nodal Protein/metabolism , Animals , Cell Differentiation , Cell Line , Germ Layers/cytology , Homeodomain Proteins/metabolism , Inhibin-beta Subunits/metabolism , Mice , Mice, Transgenic , Nanog Homeobox Protein , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/metabolism , Smad2 Protein/metabolism , Smad3 Protein/metabolism
18.
Neuron ; 82(3): 560-72, 2014 May 07.
Article in English | MEDLINE | ID: mdl-24726381

ABSTRACT

Prenatal exposure of the developing brain to various environmental challenges increases susceptibility to late onset of neuropsychiatric dysfunction; still, the underlying mechanisms remain obscure. Here we show that exposure of embryos to a variety of environmental factors such as alcohol, methylmercury, and maternal seizure activates HSF1 in cerebral cortical cells. Furthermore, Hsf1 deficiency in the mouse cortex exposed in utero to subthreshold levels of these challenges causes structural abnormalities and increases seizure susceptibility after birth. In addition, we found that human neural progenitor cells differentiated from induced pluripotent stem cells derived from schizophrenia patients show higher variability in the levels of HSF1 activation induced by environmental challenges compared to controls. We propose that HSF1 plays a crucial role in the response of brain cells to prenatal environmental insults and may be a key component in the pathogenesis of late-onset neuropsychiatric disorders.


Subject(s)
Brain Diseases/physiopathology , DNA-Binding Proteins/physiology , Environmental Exposure , Neurons/physiology , Prenatal Exposure Delayed Effects/physiopathology , Transcription Factors/physiology , Animals , Brain Diseases/embryology , Brain Diseases/etiology , Ethanol/toxicity , Female , Fetus/drug effects , Fetus/physiopathology , Heat Shock Transcription Factors , Humans , Methylmercury Compounds/toxicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Neurons/pathology , Organ Culture Techniques , Pregnancy , Prenatal Exposure Delayed Effects/etiology , Seizures/complications , Seizures/physiopathology
19.
Int J Biochem Cell Biol ; 44(10): 1593-612, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22750029

ABSTRACT

The members of the small heat shock protein (sHSP) family are molecular chaperones that play major roles in development, stress responses, and diseases, and have been envisioned as targets for therapy, particularly in cancer. The molecular mechanisms that regulate their transcription, in normal, stress, or pathological conditions, are characterized by extreme complexity and subtlety. Although historically linked to the heat shock transcription factors (HSFs), the stress-induced or developmental expression of the diverse members, including HSPB1/Hsp27/Hsp25, αA-crystallin/HSPB4, and αB-crystallin/HSPB5, relies on the combinatory effects of many transcription factors. Coupled with remarkably different cis-element architectures in the sHsp regulatory regions, they confer to each member its developmental expression or stress-inducibility. For example, multiple regulatory pathways coordinate the spatio-temporal expression of mouse αA-, αB-crystallin, and Hsp25 genes during lens development, through the action of master genes, like the large Maf family proteins and Pax6, but also HSF4. The inducibility of Hsp27 and αB-crystallin transcription by various stresses is exerted by HSF-dependent mechanisms, by which concomitant induction of Hsp27 and αB-crystallin expression is observed. In contrast, HSF-independent pathways can lead to αB-crystallin expression, but not to Hsp27 induction. Not surprisingly, deregulation of the expression of sHSP is associated with various pathologies, including cancer, neurodegenerative, or cardiac diseases. However, many questions remain to be addressed, and further elucidation of the developmental mechanisms of sHsp gene transcription might help to unravel the tissue- and stage-specific functions of this fascinating class of proteins, which might prove to be crucial for future therapeutic strategies. This article is part of a Directed Issue entitled: Small HSPs in physiology and pathology.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation, Developmental , Heat-Shock Proteins, Small/genetics , Transcription Factors/physiology , Animals , Base Sequence , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Consensus Sequence , DNA-Binding Proteins/metabolism , Heat Shock Transcription Factors , Heat-Shock Proteins, Small/metabolism , Humans , Molecular Sequence Data , Neoplasms/genetics , Neoplasms/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Response Elements , Stress, Physiological , Transcription Factors/metabolism , Transcription, Genetic
20.
Mol Biol Cell ; 22(19): 3571-83, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21813737

ABSTRACT

Heat shock response is characterized by the induction of heat shock proteins (HSPs), which facilitate protein folding, and non-HSP proteins with diverse functions, including protein degradation, and is regulated by heat shock factors (HSFs). HSF1 is a master regulator of HSP expression during heat shock in mammals, as is HSF3 in avians. HSF2 plays roles in development of the brain and reproductive organs. However, the fundamental roles of HSF2 in vertebrate cells have not been identified. Here we find that vertebrate HSF2 is activated during heat shock in the physiological range. HSF2 deficiency reduces threshold for chicken HSF3 or mouse HSF1 activation, resulting in increased HSP expression during mild heat shock. HSF2-null cells are more sensitive to sustained mild heat shock than wild-type cells, associated with the accumulation of ubiquitylated misfolded proteins. Furthermore, loss of HSF2 function increases the accumulation of aggregated polyglutamine protein and shortens the lifespan of R6/2 Huntington's disease mice, partly through αB-crystallin expression. These results identify HSF2 as a major regulator of proteostasis capacity against febrile-range thermal stress and suggest that HSF2 could be a promising therapeutic target for protein-misfolding diseases.


Subject(s)
Brain/growth & development , Heat-Shock Proteins/genetics , Heat-Shock Response/genetics , Peptides/metabolism , Transcription Factors/genetics , Animals , Brain/metabolism , Chickens , Gene Expression Regulation , Heat-Shock Proteins/deficiency , Heat-Shock Proteins/metabolism , Humans , Huntingtin Protein , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Peptides/chemistry , Protein Folding , Proteolysis , Proteostasis Deficiencies/genetics , Proteostasis Deficiencies/metabolism , Transcription Factors/deficiency , Transcription Factors/metabolism , alpha-Crystallin B Chain/genetics , alpha-Crystallin B Chain/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...