Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Cell Stress Chaperones ; 29(1): 143-157, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38311120

ABSTRACT

Preserving and regulating cellular homeostasis in the light of changing environmental conditions or developmental processes is of pivotal importance for single cellular and multicellular organisms alike. To counteract an imbalance in cellular homeostasis transcriptional programs evolved, called the heat shock response, unfolded protein response, and integrated stress response, that act cell-autonomously in most cells but in multicellular organisms are subjected to cell-nonautonomous regulation. These transcriptional programs downregulate the expression of most genes but increase the expression of heat shock genes, including genes encoding molecular chaperones and proteases, proteins involved in the repair of stress-induced damage to macromolecules and cellular structures. Sixty-one years after the discovery of the heat shock response by Ferruccio Ritossa, many aspects of stress biology are still enigmatic. Recent progress in the understanding of stress responses and molecular chaperones was reported at the 12th International Symposium on Heat Shock Proteins in Biology, Medicine and the Environment in the Old Town Alexandria, VA, USA from 28th to 31st of October 2023.


Subject(s)
Heat-Shock Proteins , Medicine , Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Heat-Shock Response/genetics , Biology
3.
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
4.
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
5.
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
6.
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
7.
Oncogene ; 38(15): 2767-2777, 2019 04.
Article in English | MEDLINE | ID: mdl-30542121

ABSTRACT

A multicenter clinical study demonstrated the presence of a loss-of-function HSP110 mutation in about 15% of colorectal cancers, which resulted from an alternative splicing and was produced at the detriment of wild-type HSP110. Patients expressing low levels of wild-type HSP110 had excellent outcomes (i.e. response to an oxaliplatin-based chemotherapy). Here, we show in vitro, in vivo, and in patients' biopsies that HSP110 co-localizes with DNA damage (γ-H2AX). In colorectal cancer cells, HSP110 translocates into the nucleus upon treatment with genotoxic chemotherapy such as oxaliplatin. Furthermore, we show that HSP110 interacts with the Ku70/Ku80 heterodimer, an essential element of the non-homologous end joining (NHEJ) repair machinery. We also demonstrate by evaluating the resolved 53BP1 foci that depletion in HSP110 impairs repair steps of the NHEJ pathway, which is associated with an increase in DNA double-strand breaks and in the cells' sensitivity to oxaliplatin. HSP110-depleted cells sensitization to oxaliplatin-induced DNA damage is abolished upon re-expression of HSP110. Confirming a role for HSP110 in DNA non-homologous repair, SCR7 and NU7026, two inhibitors of the NHEJ pathway, circumvents HSP110-induced resistance to chemotherapy. In conclusion, HSP110 through its interaction with the Ku70/80 heterodimer may participate in DNA repair, thereby inducing a protection against genotoxic therapy.


Subject(s)
Cell Nucleus/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , DNA End-Joining Repair/genetics , HSP110 Heat-Shock Proteins/genetics , Mutagens/pharmacology , Translocation, Genetic/genetics , Animals , Cell Line, Tumor , Cell Nucleus/drug effects , DNA Breaks, Double-Stranded/drug effects , DNA Damage/drug effects , DNA Damage/genetics , DNA End-Joining Repair/drug effects , DNA-Binding Proteins/genetics , HCT116 Cells , Humans , Ku Autoantigen/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Oxaliplatin/pharmacology , Translocation, Genetic/drug effects
8.
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
9.
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
10.
J Natl Cancer Inst ; 108(3)2016 Mar.
Article in English | MEDLINE | ID: mdl-26598503

ABSTRACT

BACKGROUND: Exosomes, via heat shock protein 70 (HSP70) expressed in their membrane, are able to interact with the toll-like receptor 2 (TLR2) on myeloid-derived suppressive cells (MDSCs), thereby activating them. METHODS: We analyzed exosomes from mouse (C57Bl/6) and breast, lung, and ovarian cancer patient samples and cultured cancer cells with different approaches, including nanoparticle tracking analysis, biolayer interferometry, FACS, and electron microscopy. Data were analyzed with the Student's t and Mann-Whitney tests. All statistical tests were two-sided. RESULTS: We showed that the A8 peptide aptamer binds to the extracellular domain of membrane HSP70 and used the aptamer to capture HSP70 exosomes from cancer patient samples. The number of HSP70 exosomes was higher in cancer patients than in healthy donors (mean, ng/mL ± SD = 3.5 ± 1.7 vs 0.17 ± 0.11, respectively, P = .004). Accordingly, all cancer cell lines examined abundantly released HSP70 exosomes, whereas "normal" cells did not. HSP70 had higher affinity for A8 than for TLR2; thus, A8 blocked HSP70/TLR2 association and the ability of tumor-derived exosomes to activate MDSCs. Treatment of tumor-bearing C57Bl/6 mice with A8 induced a decrease in the number of MDSCs in the spleen and inhibited tumor progression (n = 6 mice per group). Chemotherapeutic agents such as cisplatin or 5FU increase the amount of HSP70 exosomes, favoring the activation of MDSCs and hampering the development of an antitumor immune response. In contrast, this MDSC activation was not observed if cisplatin or 5FU was combined with A8. As a result, the antitumor effect of the drugs was strongly potentiated. CONCLUSIONS: A8 might be useful for quantifying tumor-derived exosomes and for cancer therapy through MDSC inhibition.


Subject(s)
Aptamers, Peptide/metabolism , Breast Neoplasms/immunology , Colonic Neoplasms/immunology , Exosomes/immunology , HSP70 Heat-Shock Proteins/metabolism , Lung Neoplasms/immunology , Myeloid Cells/immunology , Ovarian Neoplasms/immunology , Toll-Like Receptor 2/metabolism , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation , Colonic Neoplasms/drug therapy , Exosomes/drug effects , Female , Humans , Interferometry/methods , Lung Neoplasms/drug therapy , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/immunology , Ovarian Neoplasms/drug therapy , Spleen
11.
Mediators Inflamm ; 2015: 970242, 2015.
Article in English | MEDLINE | ID: mdl-26549943

ABSTRACT

Myeloproliferative neoplasms (MPN) are clonal stem cell disorders that lead to the excessive production of one or more blood cell lineages. It has been reported that, in most MPN, inflammatory cytokines are frequently increased, indicating that inflammation plays a crucial role in these disorders. Heat shock proteins (HSP) are induced in response to many stressful conditions from heat shock to hypoxia and inflammation. Besides their chaperone and cytoprotective functions, HSPs are key players during inflammation, hence the term "chaperokine." Through their chaperone activity, HSP90, a stabilizer of many oncogenes (e.g., JAK2), and HSP70, a powerful antiapoptotic chaperone, tightly regulate Nuclear Factor-kappa B signalling, a critical pathway in mediating inflammatory responses. In light of this potential, several HSP90 inhibitors have been generated as anticancer agents able to degrade oncogenes. As it turns out, however, these drugs are also potent inhibitors of the inflammatory response in various diseases. Given the chaperone potential of HSP70 and the fact that HSP90 inhibitors induce HSP70, interest in HSP70 inhibitors is also increasing. Here, we focus on the implication of HSP90 and HSP70 in inflammatory responses and on the emergence of new therapeutic approaches in MPN based on HSP inhibitors.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Inflammation Mediators/metabolism , Myeloproliferative Disorders/etiology , HSP70 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Inflammation Mediators/antagonists & inhibitors , Models, Biological , Molecular Targeted Therapy , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/metabolism , Signal Transduction
12.
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
13.
Gastroenterology ; 146(2): 401-11.e1, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24512910

ABSTRACT

BACKGROUND & AIMS: Patients with colorectal tumors with microsatellite instability (MSI) have better prognoses than patients with tumors without MSI, but have a poor response to 5-fluorouracil­based chemotherapy. A dominant-negative form of heat shock protein (HSP)110 (HSP110DE9) expressed by cancer cells with MSI, via exon skipping caused by somatic deletions in the T(17) intron repeat, sensitizes the cells to 5-fluorouracil and oxaliplatin.We investigated whether HSP110 T(17) could be used to identify patients with colorectal cancer who would benefit from adjuvant chemotherapy with 5-fluorouracil and oxaliplatin. METHODS: We characterized the interaction between HSP110 and HSP110DE9 using surface plasmon resonance. By using polymerase chain reaction and fragment analysis, we examined how the size of somatic allelic deletions in HSP110 T(17) affected the HSP110 protein expressed by tumor cells. We screened 329 consecutive patients with stage II­III colorectal tumors with MSI who underwent surgical resection at tertiary medical centers for HSP110 T(17). RESULTS: HSP110 and HSP110DE9 interacted in a1:1 ratio. Tumor cells with large deletions in T(17) had increased ratios of HSP110DE9:HSP110, owing to the loss of expression of full-length HSP110. Deletions in HSP110 T(17) were mostly biallelic in primary tumor samples with MSI. Patients with stage II­III cancer who received chemotherapy and had large HSP110 T(17) deletions (≥5 bp; 18 of 77 patients, 23.4%) had longer times of relapse-free survival than patients with small or no deletions (≤4 bp; 59 of 77 patients, 76.6%) in multivariate analysis (hazard ratio, 0.16; 95% confidence interval, 0.012­0.8; P = .03). We found a significant interaction between chemotherapy and T17 deletion (P =.009). CONCLUSIONS: About 25% of patients with stages II­III colorectal tumors with MSI have an excellent response to chemotherapy, due to large, biallelic deletions in the T(17) intron repeat of HSP110 in tumor DNA.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Base Sequence , Biomarkers, Tumor/genetics , Colorectal Neoplasms/drug therapy , HSP110 Heat-Shock Proteins/genetics , Microsatellite Instability , Sequence Deletion , Aged , Antineoplastic Agents/administration & dosage , Biomarkers, Tumor/chemistry , Biomarkers, Tumor/metabolism , Blotting, Western , Cell Line, Tumor , Chemotherapy, Adjuvant , Colectomy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Colorectal Neoplasms/surgery , Female , Fluorouracil/administration & dosage , Follow-Up Studies , HSP110 Heat-Shock Proteins/chemistry , HSP110 Heat-Shock Proteins/metabolism , Humans , Introns , Leucovorin/administration & dosage , Male , Models, Molecular , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Retrospective Studies , Surface Plasmon Resonance , Survival Analysis , Treatment Outcome
14.
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
15.
Blood ; 119(6): 1532-42, 2012 Feb 09.
Article in English | MEDLINE | ID: mdl-22160620

ABSTRACT

Normal human erythroid cell maturation requests the transcription factor GATA-1 and a transient activation of caspase-3, with GATA-1 being protected from caspase-3-mediated cleavage by interaction with the chaperone heat shock protein 70 (Hsp70) in the nucleus. Erythroid cell dysplasia observed in early myelodysplastic syndromes (MDS) involves impairment of differentiation and excess of apoptosis with a burst of caspase activation. Analysis of gene expression in MDS erythroblasts obtained by ex vivo cultures demonstrates the down-regulation of a set of GATA-1 transcriptional target genes, including GYPA that encodes glycophorin A (GPA), and the up-regulation of members of the HSP70 family. GATA-1 protein expression is decreased in MDS erythroblasts, but restores in the presence of a pan-caspase inhibitor. Expression of a mutated GATA-1 that cannot be cleaved by caspase-3 rescues the transcription of GATA-1 targets, and the erythroid differentiation, but does not improve survival. Hsp70 fails to protect GATA-1 from caspases because the protein does not accumulate in the nucleus with active caspase-3. Expression of a nucleus-targeted mutant of Hsp70 protects GATA-1 and rescues MDS erythroid cell differentiation. Alteration of Hsp70 cytosolic-nuclear shuttling is a major feature of MDS that favors GATA-1 cleavage and differentiation impairment, but not apoptosis, in dysplastic erythroblasts.


Subject(s)
Cell Nucleus/metabolism , Erythropoiesis/genetics , GATA1 Transcription Factor/genetics , HSP70 Heat-Shock Proteins/genetics , Adult , Aged , Aged, 80 and over , Caspase 3/genetics , Caspase 3/metabolism , Cell Differentiation/genetics , Cells, Cultured , Erythroblasts/metabolism , Erythroid Cells/metabolism , Female , GATA1 Transcription Factor/metabolism , Gene Expression Profiling , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HSP70 Heat-Shock Proteins/metabolism , Humans , Immunoblotting , Male , Microscopy, Fluorescence , Middle Aged , Mutation , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , U937 Cells
16.
Nat Med ; 17(10): 1283-9, 2011 Sep 25.
Article in English | MEDLINE | ID: mdl-21946539

ABSTRACT

Heat shock proteins (HSPs) are necessary for cancer cell survival. We identified a mutant of HSP110 (HSP110ΔE9) in colorectal cancer showing microsatellite instability (MSI CRC), generated from an aberrantly spliced mRNA and lacking the HSP110 substrate-binding domain. This mutant was expressed at variable levels in almost all MSI CRC cell lines and primary tumors tested. HSP110ΔE9 impaired both the normal cellular localization of HSP110 and its interaction with other HSPs, thus abrogating the chaperone activity and antiapoptotic function of HSP110 in a dominant-negative manner. HSP110ΔE9 overexpression caused the sensitization of cells to anticancer agents such as oxaliplatin and 5-fluorouracil, which are routinely prescribed in the adjuvant treatment of people with CRC. The survival and response to chemotherapy of subjects with MSI CRCs was associated with the tumor expression level of HSP110ΔE9. HSP110 may thus constitute a major determinant for both prognosis and treatment response in CRC.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , HSP110 Heat-Shock Proteins/metabolism , Antineoplastic Agents/therapeutic use , Blotting, Western , Cell Line, Tumor , Colorectal Neoplasms/genetics , DNA Primers/genetics , Fluorescent Antibody Technique , Fluorouracil , HSP110 Heat-Shock Proteins/genetics , Humans , Immunoprecipitation , Microsatellite Instability , Mutation/genetics , Organoplatinum Compounds , Oxaliplatin , Plasmids/genetics , Prognosis , Real-Time Polymerase Chain Reaction , Regression Analysis , Transfection
17.
Cancer Res ; 71(2): 484-95, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21224349

ABSTRACT

The inhibition of heat shock protein 70 (HSP70) is an emerging strategy in cancer therapy. Unfortunately, no specific inhibitors are clinically available. By yeast two-hybrid screening, we have identified multiple peptide aptamers that bind HSP70. When expressed in human tumor cells, two among these peptide aptamers-A8 and A17-which bind to the peptide-binding and the ATP-binding domains of HSP70, respectively, specifically inhibited the chaperone activity, thereby increasing the cells' sensitivity to apoptosis induced by anticancer drugs. The 13-amino acid peptide from the variable region of A17 (called P17) retained the ability to specifically inhibit HSP70 and induced the regression of subcutaneous tumors in vivo after local or systemic injection. This antitumor effect was associated with an important recruitment of macrophages and T lymphocytes into the tumor bed. Altogether, these data indicate that peptide aptamers or peptides that target HSP70 may be considered as novel lead compounds for cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Aptamers, Peptide/pharmacology , HSP70 Heat-Shock Proteins/antagonists & inhibitors , Molecular Targeted Therapy/methods , Peptides/pharmacology , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Aptamers, Peptide/chemistry , Aptamers, Peptide/genetics , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , HeLa Cells , Humans , Melanoma, Experimental/drug therapy , Mice , Mice, Inbred C57BL , Peptides/chemistry , Peptides/genetics , Protein Structure, Tertiary , Rats , Transfection
18.
Cancers (Basel) ; 3(1): 1158-81, 2011 Mar 07.
Article in English | MEDLINE | ID: mdl-24212658

ABSTRACT

Heat Shock Factors (HSF) form a family of transcription factors (four in mammals) which were named according to the discovery of their activation by a heat shock. HSFs trigger the expression of genes encoding Heat Shock Proteins (HSPs) that function as molecular chaperones, contributing to establish a cytoprotective state to various proteotoxic stresses and in pathological conditions. Increasing evidence indicates that this ancient transcriptional protective program acts genome-widely and performs unexpected functions in the absence of experimentally defined stress. Indeed, HSFs are able to re-shape cellular pathways controlling longevity, growth, metabolism and development. The most well studied HSF, HSF1, has been found at elevated levels in tumors with high metastatic potential and is associated with poor prognosis. This is partly explained by the above-mentioned cytoprotective (HSP-dependent) function that may enable cancer cells to adapt to the initial oncogenic stress and to support malignant transformation. Nevertheless, HSF1 operates as major multifaceted enhancers of tumorigenesis through, not only the induction of classical heat shock genes, but also of "non-classical" targets. Indeed, in cancer cells, HSF1 regulates genes involved in core cellular functions including proliferation, survival, migration, protein synthesis, signal transduction, and glucose metabolism, making HSF1 a very attractive target in cancer therapy. In this review, we describe the different physiological roles of HSFs as well as the recent discoveries in term of non-cogenic potential of these HSFs, more specifically associated to the activation of "non-classical" HSF target genes. We also present an update on the compounds with potent HSF1-modulating activity of potential interest as anti-cancer therapeutic agents.

19.
Blood ; 116(1): 85-96, 2010 Jul 08.
Article in English | MEDLINE | ID: mdl-20410505

ABSTRACT

Heat shock protein 27 (HSP27) is a chaperone whose cellular expression increases in response to various stresses and protects the cell either by inhibiting apoptotic cell death or by promoting the ubiquitination and proteasomal degradation of specific proteins. Here, we show that globin transcription factor 1 (GATA-1) is a client protein of HSP27. In 2 models of erythroid differentiation; that is, in the human erythroleukemia cell line, K562 induced to differentiate into erythroid cells on hemin exposure and CD34(+) human cells ex vivo driven to erythroid differentiation in liquid culture, depletion of HSP27 provokes an accumulation of GATA-1 and impairs terminal maturation. More specifically, we demonstrate that, in the late stages of the erythroid differentiation program, HSP27 is phosphorylated in a p38-dependent manner, enters the nucleus, binds to GATA-1, and induces its ubiquitination and proteasomal degradation, provided that the transcription factor is acetylated. We conclude that HSP27 plays a role in the fine-tuning of terminal erythroid differentiation through regulation of GATA-1 content and activity.


Subject(s)
Cell Differentiation , Erythroid Cells/metabolism , GATA1 Transcription Factor/metabolism , HSP27 Heat-Shock Proteins/metabolism , Animals , Antigens, CD34/blood , COS Cells , Cell Nucleus/metabolism , Cells, Cultured , Chlorocebus aethiops , Erythroid Cells/cytology , Erythroid Cells/drug effects , GATA1 Transcription Factor/genetics , HSP27 Heat-Shock Proteins/genetics , HeLa Cells , Heat-Shock Proteins , Humans , Imidazoles/pharmacology , Immunoblotting , Interleukin-6/pharmacology , K562 Cells , Leupeptins/pharmacology , Molecular Chaperones , Phosphorylation/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , Protein Binding , Pyridines/pharmacology , RNA Interference , Transforming Growth Factor beta/pharmacology , Ubiquitination/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
20.
Mol Biol Cell ; 21(8): 1423-34, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20200223

ABSTRACT

Atypical protein kinase Czeta (PKCzeta) is emerging as a mediator of differentiation. Here, we describe a novel role for PKCzeta in myogenic differentiation, demonstrating that PKCzeta activity is indispensable for differentiation of both C2C12 and mouse primary myoblasts. PKCzeta was found to be associated with and to regulate the Cdk5/p35 signaling complex, an essential factor for both neuronal and myogenic differentiation. Inhibition of PKCzeta activity prevented both myotube formation and simultaneous reorganization of the nestin intermediate filament cytoskeleton, which is known to be regulated by Cdk5 during myogenesis. p35, the Cdk5 activator, was shown to be a specific phosphorylation target of PKCzeta. PKCzeta-mediated phosphorylation of Ser-33 on p35 promoted calpain-mediated cleavage of p35 to its more active and stable fragment, p25. Strikingly, both calpain activation and the calpain-mediated cleavage of p35 were shown to be PKCzeta-dependent in differentiating myoblasts. Overall, our results identify PKCzeta as a controller of myogenic differentiation by its regulation of the phosphorylation-dependent and calpain-mediated p35 cleavage, which is crucial for the amplification of the Cdk5 activity that is required during differentiation.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Muscle Development , Protein Kinase C/metabolism , Signal Transduction , Animals , COS Cells , Calpain/metabolism , Cell Differentiation/drug effects , Chlorocebus aethiops , Intermediate Filament Proteins/metabolism , Mice , Models, Biological , Muscle Development/drug effects , Myoblasts/cytology , Myoblasts/drug effects , Myoblasts/enzymology , Nerve Tissue Proteins/metabolism , Nestin , Phosphorylation/drug effects , Phosphotransferases/metabolism , Protein Binding/drug effects , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...