Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 21
1.
Reproduction ; 158(2): R49-R65, 2019 08.
Article En | MEDLINE | ID: mdl-30978695

The mammalian zygote is a totipotent cell that generates all the cells of a new organism through embryonic development. However, if one asks about the totipotency of blastomeres after one or two zygotic divisions, opinions differ. As it is impossible to determine the individual developmental potency of early blastomeres in an intact embryo, experiments of blastomere isolation were conducted in various species, showing that two-cell blastomeres could give rise to a new organism when sister cells were separated. A mainstream interpretation was that each of the sister mammalian blastomeres was equally totipotent. However, reevaluation of those experiments raised some doubts about the real prevalence of cases in which this interpretation could truly be validated. We compiled experiments that tested the individual developmental potency of early mammalian blastomeres in a cell-autonomous way (i.e. excluding nuclear transfer and chimera production). We then confronted the developmental abilities with reported molecular differences between sister blastomeres. The reevaluated observations were at odds with the mainstream view: A viable two-cell embryo can already include one non-totipotent blastomere. We were, thus, led to propose a revised model for totipotency continuity based on the construction of the zygote as a mosaic, which accounts for differential inheritance of totipotency-relevant components between sister blastomeres. This takes place with no preordained mechanisms that would ensure a reproducible partition. This model, which is compatible with the body of data on regulative properties of mammalian early embryos, aims at tempering the rigid interpretation that discounted maternal constraints on totipotency.


Blastomeres/physiology , Models, Biological , Zygote/physiology , Animals , Humans , Mosaicism
2.
Adv Anat Embryol Cell Biol ; 222: 1-27, 2017.
Article En | MEDLINE | ID: mdl-28389748

Early embryos develop from fertilized eggs using materials that are stored during oocyte growth and which can be defined as maternal contribution (molecules, factors, or determinants). Several heat shock proteins (HSPs) and the heat shock transcriptional factor (HSF) are part of the maternal contribution that is critical for successful embryogenesis and reproduction. A maternal role for heat shock-related genes was mainly demonstrated in genetic experimental organisms (e.g., fly, nematode, mouse). Nowadays, an increasing number of "omics" data are produced from a large panel of organisms implementing a catalog of maternal and/or embryonic HSPs and HSFs. However, for most of them, it remains to better understand their potential roles in this context. Existing and future genome-wide screens mainly set up to create loss-of-function are likely to improve this situation. This chapter will discuss available data from various experimental organisms following the developmental steps from egg production to early embryogenesis.


Embryonic Development/genetics , Heat-Shock Proteins/genetics , Oogenesis/genetics , Animals , Gene Expression Regulation, Developmental/genetics , Humans , Zygote/metabolism
3.
J Biol Chem ; 291(29): 14939-53, 2016 07 15.
Article En | MEDLINE | ID: mdl-27226619

Mutations of HSPB5 (also known as CRYAB or αB-crystallin), a bona fide heat shock protein and molecular chaperone encoded by the HSPB5 (crystallin, alpha B) gene, are linked to multisystem disorders featuring variable combinations of cataracts, cardiomyopathy, and skeletal myopathy. This study aimed to investigate the pathological mechanisms involved in an early-onset myofibrillar myopathy manifesting in a child harboring a homozygous recessive mutation in HSPB5, 343delT. To study HSPB5 343delT protein dynamics, we utilize model cell culture systems including induced pluripotent stem cells derived from the 343delT patient (343delT/343delT) along with isogenic, heterozygous, gene-corrected control cells (WT KI/343delT) and BHK21 cells, a cell line lacking endogenous HSPB5 expression. 343delT/343delT and WT KI/343delT-induced pluripotent stem cell-derived skeletal myotubes and cardiomyocytes did not express detectable levels of 343delT protein, contributable to the extreme insolubility of the mutant protein. Overexpression of HSPB5 343delT resulted in insoluble mutant protein aggregates and induction of a cellular stress response. Co-expression of 343delT with WT prevented visible aggregation of 343delT and improved its solubility. Additionally, in vitro refolding of 343delT in the presence of WT rescued its solubility. We demonstrate an interaction between WT and 343delT both in vitro and within cells. These data support a loss-of-function model for the myopathy observed in the patient because the insoluble mutant would be unavailable to perform normal functions of HSPB5, although additional gain-of-function effects of the mutant protein cannot be excluded. Additionally, our data highlight the solubilization of 343delT by WT, concordant with the recessive inheritance of the disease and absence of symptoms in carrier individuals.


Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cataract/genetics , Cataract/metabolism , Muscular Diseases/genetics , Muscular Diseases/metabolism , alpha-Crystallin B Chain/genetics , alpha-Crystallin B Chain/metabolism , Cardiomyopathies/etiology , Cataract/etiology , Female , Homozygote , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Models, Biological , Muscle Fibers, Skeletal/metabolism , Muscular Diseases/etiology , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Myocytes, Cardiac/metabolism , Pedigree , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Deletion , Solubility , alpha-Crystallin B Chain/chemistry
4.
Curr Protein Pept Sci ; 15(3): 189-204, 2014 May.
Article En | MEDLINE | ID: mdl-24694370

Cardiomyocytes are best known for their spontaneous beating activity, large cell size, and low regenerative capacity during adulthood. The mechanical activity of cardiomyocytes depends on a sophisticated contractile apparatus comprised of sarcomeres whose rhythmic contraction relies on Ca(2+) transients with a high level of energy consumption. Hence the proper folding and assembly of the sarcomeric and other accessory proteins involved in those diverse functions (i.e., structural, mechanical, energy exchange and production) is critical for muscle mechanics. Chaperone proteins assist other polypeptides to reach their proper conformation, activity and/or location. Consequently, chaperone-like functions are important for the healthy heart but assume greater relevance during cardiac diseases when such chaperone proteins are recruited: 1) to protect cardiac cells against adverse effects during the pathological transition, and 2) to mitigate certain pathogenic mechanisms per se. Protein misfolding is observed as a consequence of inappropriate intracellular environment with acquired conditions (e.g., ischemia/reperfusion and redox imbalance) or because of mutations, which can modify primary to quaternary protein structures. In this review, we discuss the importance of cardiac chaperones while emphasizing the genetic origin (modification of gene/protein sequence) of cardiac protein misfolding and their consequences on the cardiomyocytes leading to organ dysfunction and failure.


Heart Diseases/metabolism , Molecular Chaperones/metabolism , Proteostasis Deficiencies/metabolism , Animals , Heart Diseases/genetics , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Proteostasis Deficiencies/genetics
5.
Antioxid Redox Signal ; 20(18): 2891-906, 2014 Jun 20.
Article En | MEDLINE | ID: mdl-24180415

AIMS: The human mutation R120G in the αB-crystallin (CRYAB) causes a multisystemic disease that is characterized by hypertrophic cardiomyopathy and cytoplasmic protein aggregates. In transgenic mice, human R120GCRYAB (hR120GTg) expression in heart sequentially modifies the REDOX status, in part by the activation of the nuclear factor, erythroid derived 2, like 2 (Nrf2). Thioredoxin system (TS) components are NRF2 target genes, so it could be hypothesized that TS was affected in hR120GTg mice. RESULTS: Transgenic hearts overexpressed thioredoxin 1 (Trx1), which was identified by isotope coded affinity tag-mass spectrometry, among hundreds of peptides displaying an increased reduced/oxidized ratio. Coupled to this higher level of reduced cysteines, the activity of thioredoxin reductase 1 (TrxR1) was augmented by 2.5-fold. Combining mutiple experimental approaches, the enzymatic regulation of TrxR1 by a histone deacetylase 3 (HDAC3)-dependent level of acetylation was confirmed. In vitro and in vivo functional tests established that TrxR1 activity is required to mitigate aggregate development, and this could be mediated by Bcl-2-associated athanogene 3 (BAG3) as a potential TS substrate. INNOVATION AND CONCLUSIONS: This study uncovers the compartmentalized changes and the involvement of TS in the cardiac stress response elicited by misfolded proteins such as R120GCRYAB. Our work suggests that R120GCRYAB triggers a defensive pathway acting through the newly identified interacting partners HDAC3, TrxR1, and BAG3 to counter aggregate growth. Therefore, those interactors may function as modifier genes contributing to the variable onset and expressivity of such human diseases. Furthermore, our work underscores the potential organismal effects of pharmacological interventions targeting TS and HDAC.


Cardiomyopathy, Hypertrophic/genetics , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Oxidation-Reduction , Protein Aggregation, Pathological/genetics , alpha-Crystallin B Chain/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Histone Deacetylases/metabolism , Humans , Isotope Labeling , Mass Spectrometry , Mice , Mice, Transgenic , NF-E2-Related Factor 2/metabolism , Thioredoxin Reductase 1/metabolism , Thioredoxins/metabolism
6.
PLoS One ; 7(8): e42118, 2012.
Article En | MEDLINE | ID: mdl-22870288

BACKGROUND: CryAB (HspB5) and HspB2, two small heat shock genes located adjacently in the vertebrate genome, are hypothesized to play distinct roles. Mice lacking both cryab and hspb2 (DKO) are viable and exhibit adult-onset degeneration of skeletal muscle but confounding results from independent groups were reported for cardiac responses to different stressful conditions (i.e., ischemia/reperfusion or pressure overload). To determine the specific requirements of HSPB2 in heart, we generated cardiac-specific HSPB2 deficient (HSPB2cKO) mice and examined their cardiac function under basal conditions and following cardiac pressure overload. METHODOLOGY/PRINCIPAL FINDINGS: Transverse aortic constriction (TAC) or sham surgery was performed in HSPB2cKO mice and their littermates (HSPB2wt mice). Eight weeks after TAC, we found that expression of several small HSPs (HSPB2, 5, 6) was not markedly modified in HSPB2wt mice. Both cardiac function and the hypertrophic response remained similar in HSPB2cKO and HSPB2wt hearts. In addition, mitochondrial respiration and ATP production assays demonstrated that the absence of HSPB2 did not change mitochondrial metabolism in basal conditions. However, fatty acid supported state 3 respiration rate (ADP stimulated) in TAC operated HSPB2cKO hearts was significantly reduced in compared with TAC operated HSPB2wt mice (10.5±2.2 vs. 12.8±2.5 nmol O(2)/min/mg dry fiber weight, P<0.05), and ATP production in HSPB2cKO hearts was significantly reduced in TAC compared with sham operated mice (29.8±0.2 vs. 21.1±1.8 nmol ATP/min/mg dry fiber weight, P<0.05). Although HSPB2 was not associated with mitochondria under cardiac stress, absence of HSPB2 led to changes in transcript levels of several metabolic and mitochondrial regulator genes. CONCLUSIONS/SIGNIFICANCE: The present study indicates that HSPB2 can be replaced by other members of the multigene small HSP family under basal conditions while HSPB2 is implicated in the regulation of metabolic/mitochondrial function under cardiac stress such pressure overload.


Adenosine Triphosphate/biosynthesis , Blood Pressure , Cardiomegaly/metabolism , HSP27 Heat-Shock Proteins , Mitochondria, Heart/metabolism , Oxygen Consumption , Adenosine Triphosphate/genetics , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Mice , Mice, Knockout , Mitochondria, Heart/genetics , Mitochondria, Heart/pathology
7.
Int J Biochem Cell Biol ; 44(10): 1632-45, 2012 Oct.
Article En | MEDLINE | ID: mdl-22710345

A timely review series on small heat shock proteins has to appropriately examine their fundamental properties and implications in the cardiovascular system since several members of this chaperone family exhibit robust expression in the myocardium and blood vessels. Due to energetic and metabolic demands, the cardiovascular system maintains a high mitochondrial activity but irreversible oxidative damage might ensue from increased production of reactive oxygen species. How equilibrium between their production and scavenging is achieved becomes paramount for physiological maintenance. For example, heat shock protein B1 (HSPB1) is implicated in maintaining this equilibrium or redox homeostasis by upholding the level of glutathione, a major redox mediator. Studies of gain or loss of function achieved by genetic manipulations have been highly informative for understanding the roles of those proteins. For example, genetic deficiency of several small heat shock proteins such as HSPB5 and HSPB2 is well-tolerated in heart cells whereas a single missense mutation causes human pathology. Such evidence highlights both the profound genetic redundancy observed among the multigene family of small heat shock proteins while underscoring the role proteotoxicity plays in driving disease pathogenesis. We will discuss the available data on small heat shock proteins in the cardiovascular system, redox metabolism and human diseases. From the medical perspective, we envision that such emerging knowledge of the multiple roles small heat shock proteins exert in the cardiovascular system will undoubtedly open new avenues for their identification and possible therapeutic targeting in humans. This article is part of a Directed Issue entitled: Small HSPs in physiology and pathology.


Cardiovascular Diseases/metabolism , Heat-Shock Proteins, Small/metabolism , Oxidative Stress , Animals , Cardiovascular Diseases/genetics , Cardiovascular System/embryology , Cardiovascular System/growth & development , Cardiovascular System/metabolism , Gene Expression Regulation, Developmental , Heat-Shock Proteins, Small/physiology , Humans , Mutation , Oxidation-Reduction , Proteostasis Deficiencies/genetics , Proteostasis Deficiencies/metabolism
8.
Am J Physiol Heart Circ Physiol ; 302(1): H24-37, 2012 Jan 01.
Article En | MEDLINE | ID: mdl-22003057

Force-generating contractile cells of the myocardium must achieve and maintain their primary function as an efficient mechanical pump over the life span of the organism. Because only half of the cardiomyocytes can be replaced during the entire human life span, the maintenance strategy elicited by cardiac cells relies on uninterrupted renewal of their components, including proteins whose specialized functions constitute this complex and sophisticated contractile apparatus. Thus cardiac proteins are continuously synthesized and degraded to ensure proteome homeostasis, also termed "proteostasis." Once synthesized, proteins undergo additional folding, posttranslational modifications, and trafficking and/or become involved in protein-protein or protein-DNA interactions to exert their functions. This includes key transient interactions of cardiac proteins with molecular chaperones, which assist with quality control at multiple levels to prevent misfolding or to facilitate degradation. Importantly, cardiac proteome maintenance depends on the cellular environment and, in particular, the reduction-oxidation (REDOX) state, which is significantly different among cardiac organelles (e.g., mitochondria and endoplasmic reticulum). Taking into account the high metabolic activity for oxygen consumption and ATP production by mitochondria, it is a challenge for cardiac cells to maintain the REDOX state while preventing either excessive oxidative or reductive stress. A perturbed REDOX environment can affect protein handling and conformation (e.g., disulfide bonds), disrupt key structure-function relationships, and trigger a pathogenic cascade of protein aggregation, decreased cell survival, and increased organ dysfunction. This review covers current knowledge regarding the general domain of REDOX state and protein folding, specifically in cardiomyocytes under normal-healthy conditions and during disease states associated with morbidity and mortality in humans.


Heart Diseases/metabolism , Muscle Proteins/metabolism , Myocardial Contraction , Myocardium/metabolism , Oxidative Stress , Animals , Heart Diseases/drug therapy , Heart Diseases/physiopathology , Homeostasis , Humans , Muscle Proteins/chemistry , Myocardial Contraction/drug effects , Oxidation-Reduction , Protein Conformation , Protein Folding , Structure-Activity Relationship
9.
Mol Cell Biol ; 31(16): 3410-23, 2011 Aug.
Article En | MEDLINE | ID: mdl-21690297

Heat shock factor 1 (HSF1), while recognized as the major regulator of the heat shock transcriptional response, also exerts important functions during mammalian embryonic development and gametogenesis. In particular, HSF1 is required for oocyte maturation, the adult phase of meiosis preceding fertilization. To identify HSF1 target genes implicated in this process, comparative transcriptomic analyses were performed with wild-type and HSF-deficient oocytes. This revealed a network of meiotic genes involved in cohesin and synaptonemal complex (SC) structures, DNA recombination, and the spindle assembly checkpoint (SAC). All of them were found to be regulated by HSF1 not only during adult but also in embryonic phases of female meiosis. Additional investigations showed that SC, recombination nodules, and DNA repair were affected in Hsf1(-/-) oocytes during prenatal meiotic prophase I. However, targeting Hsf1 deletion to postnatal oocytes (using Zp3 Cre; Hsf1(loxP/loxP)) did not fully rescue the chromosomal anomalies identified during meiotic maturation, which possibly caused a persistent SAC activation. This would explain the metaphase I arrest previously described in HSF1-deficient oocytes since SAC inhibition circumvented this block. This work provides new insights into meiotic gene regulation and points out potential links between cellular stress and the meiotic anomalies frequently observed in humans.


DNA-Binding Proteins/genetics , Embryonic Development , Gametogenesis/genetics , Gene Expression Regulation/physiology , Meiosis , Transcription Factors/genetics , Age Factors , Animals , Embryonic Development/genetics , Female , Gene Expression Profiling , Gene Regulatory Networks , Heat Shock Transcription Factors , Mice , Mice, Knockout , Oocytes
10.
PLoS One ; 6(2): e17109, 2011 Feb 15.
Article En | MEDLINE | ID: mdl-21358806

BACKGROUND: Hsp90b1 is an endoplasmic reticulum (ER) chaperone (also named Grp94, ERp99, gp96,Targ2, Tra-1, Tra1, Hspc4) (MGI:98817) contributing with Hspa5 (also named Grp78, BIP) (MGI:95835) to protein folding in ER compartment. Besides its high protein expression in mouse oocytes, little is known about Hsp90b1 during the transition from oocyte-to-embryo. Because the constitutive knockout of Hsp90b1 is responsible for peri-implantation embryonic lethality, it was not yet known whether Hsp90b1 is a functionally important maternal factor. METHODOLOGY/FINDINGS: To circumvent embryonic lethality, we established an oocyte-specific conditional knockout line taking advantage of the more recently created floxed Hsp90b1 line (Hsp90b1(flox), MGI:3700023) in combination with the transgenic mouse line expressing the cre recombinase under the control of zona pellucida 3 (ZP3) promoter (Zp3-cre, MGI:2176187). Altered expression of Hsp90b1 in growing oocytes provoked a limited, albeit significant reduction of the zona pellucida thickness but no obvious anomalies in follicular growth, meiotic maturation or fertilization. Interestingly, mutant zygotes obtained from oocytes lacking Hsp90b1 were unable to reach the 2-cell stage. They exhibited either a G2/M block or, more frequently an abnormal mitotic spindle leading to developmental arrest. Despite the fact that Hspa5 displayed a similar profile of expression as Hsp90b1, we found that HSPA5 and HSP90B1 did not fully colocalize in zygotes suggesting distinct function for the two chaperones. Consequently, even if HSPA5 was overexpressed in Hsp90b1 mutant embryos, it did not compensate for HSP90B1 deficiency. Finally, further characterization of ER compartment and cytoskeleton revealed a defective organization of the cytoplasmic region surrounding the mutant zygotic spindle. CONCLUSIONS: Our findings demonstrate that the maternal contribution of Hsp90b1 is critical for the development of murine zygotes. All together our data indicate that Hsp90b1 is involved in unique and specific aspects of the first mitosis, which brings together the maternal and paternal genomes on a single spindle.


HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/physiology , Mitosis/genetics , Oocytes/metabolism , Zygote/physiology , Animals , Animals, Newborn , Cells, Cultured , Embryo, Mammalian , Endoplasmic Reticulum Chaperone BiP , Female , Gene Deletion , Gene Expression Regulation, Developmental/physiology , HSP90 Heat-Shock Proteins/metabolism , Mice , Mice, Transgenic , Models, Biological , Mutagenesis, Site-Directed , Oogenesis/genetics , Oogenesis/physiology , Organ Specificity/genetics , Time Factors , Zygote/metabolism
11.
J Biol Chem ; 285(45): 34469-76, 2010 Nov 05.
Article En | MEDLINE | ID: mdl-20802198

Heat shock factor 1 (HSF1) is an important transcription factor in cellular stress responses, cancer, aging, and developmental processes including gametogenesis. Disruption of Hsf1, together with another HSF family member, Hsf2, causes male sterility and complete lack of mature sperm in mice, but the specific role of HSF1 in spermatogenesis has remained unclear. Here, we show that HSF1 is transiently expressed in meiotic spermatocytes and haploid round spermatids in mouse testis. The Hsf1(-/-) male mice displayed regions of seminiferous tubules containing only spermatogonia and increased morphological abnormalities in sperm heads. In search for HSF1 target genes, we identified 742 putative promoters in mouse testis. Among them, the sex chromosomal multicopy genes that are expressed in postmeiotic cells were occupied by HSF1. Given that the sex chromatin mostly is repressed during and after meiosis, it is remarkable that HSF1 directly regulates the transcription of sex-linked multicopy genes during postmeiotic repression. In addition, our results show that HSF1 localizes to the sex body prior to the meiotic divisions and to the sex chromocenter after completed meiosis. To the best of our knowledge, HSF1 is the first known transcription factor found at the repressed sex chromatin during meiosis.


DNA-Binding Proteins/metabolism , Meiosis/physiology , Seminiferous Tubules/metabolism , Sex Chromatin/metabolism , Spermatogenesis/physiology , Spermatozoa/metabolism , Transcription Factors/metabolism , Animals , DNA-Binding Proteins/genetics , Female , Gene Expression Regulation/physiology , Heat Shock Transcription Factors , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Infertility, Male/genetics , Infertility, Male/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Multigene Family/physiology , Sex Chromatin/genetics , Transcription Factors/genetics
12.
J Biol Chem ; 284(14): 9521-8, 2009 Apr 03.
Article En | MEDLINE | ID: mdl-19158073

Heat shock transcription factor 1 (HSF1) is the main regulator of the stress response that triggers the transcription of several genes encoding heat shock proteins (Hsps). Hsps act as molecular chaperones involved in protein folding, stability, and trafficking. HSF1 is highly expressed in oocytes and Hsf1 knock-out in mice revealed that in the absence of stress this factor plays an important role in female reproduction. We previously reported that Hsf1(-/-) females produce oocytes but no viable embryos. Consequently, we asked whether oocytes require HSF1 to regulate a particular set of Hsps necessary for them to develop. We find that Hsp90alpha (Hspaa1) is the major HSF1-dependent chaperone inasmuch as Hsf1 knock-out resulted in Hsp90-depleted oocytes. These oocytes exhibited delayed germinal vesicle breakdown (or G(2)/M transition), partial meiosis I block, and defective asymmetrical division. To probe the role of Hsp90alpha in this meiotic syndrome, we analyzed meiotic maturation in wild-type oocytes treated with a specific inhibitor of Hsp90, 17-allylamino-17-demethoxy-geldanamycin, and observed similar defects. At the molecular level we showed that, together with these developmental anomalies, CDK1 and MAPK, key meiotic kinases, were significantly disturbed. Thus, our data demonstrate that HSF1 is a maternal transcription factor essential for normal progression of meiosis.


DNA-Binding Proteins/metabolism , Gene Expression Regulation , HSP90 Heat-Shock Proteins/metabolism , Meiosis , Oocytes/cytology , Oocytes/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , Cell Differentiation , Cytoplasm/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Female , HSP90 Heat-Shock Proteins/genetics , Heat Shock Transcription Factors , MAP Kinase Signaling System , Mice , Mice, Knockout , Protein Isoforms/genetics , Protein Isoforms/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics
13.
Biol Reprod ; 79(6): 1092-101, 2008 Dec.
Article En | MEDLINE | ID: mdl-18703420

Heat-shock factor 1 (HSF1) protects cells and organisms against various types of stress, either by triggering a complex response that promotes cell survival or by triggering cell death when stress-induced alterations cannot be rescued. Although this dual role of HSF1 was observed in spermatogenesis exposed to heat shock or proteotoxic stress, HSF1 was also reported to contribute to cell resistance against genotoxic stress, such as that caused by doxorubicin, an anticancer drug in common clinical use. To better understand the stress/cell-dependent functions of HSF1, we used wild-type and Hsf1(tm1Ijb)/Hsf1(tm1Ijb) males to determine the role of HSF1 in the genotoxic stress response elicited in spermatogenic cells. Within 2 days after a single intraperitoneal injection of doxorubicin (DOXO; 5 mg/kg), proliferation of Hsf1+/+ but not Hsf1-/- spermatogenic cells was significantly reduced, whereas cell death was increased in mitotic germ cells and metaphase I spermatocytes. By 21 days, meiotic cells were depleted in all treated Hsf1+/+ testes but not in Hsf1-/- ones. Nevertheless, after 3 mo, spermatogenesis showed better signs of recovery in Hsf1+/+ than in Hsf1-/- males. Taken together, these data indicate that acute response to genotoxic stress in the testis involves HSF1-dependent mechanisms that induce apoptotic cell death in a TRP53-independent manner, but also intervene on a longer term to restore seminiferous tubules.


Antibiotics, Antineoplastic/toxicity , DNA-Binding Proteins/physiology , Doxorubicin/toxicity , Mutagenesis/drug effects , Mutagens/toxicity , Testis/drug effects , Testis/physiology , Transcription Factors/physiology , Animals , Antimetabolites , Blotting, Western , Bromodeoxyuridine , DNA-Binding Proteins/genetics , Heat Shock Transcription Factors , Immunohistochemistry , In Situ Nick-End Labeling , Male , Meiosis/drug effects , Meiosis/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Proliferating Cell Nuclear Antigen/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sperm Count , Sperm Head/drug effects , Sperm Head/physiology , Spermatogenesis/drug effects , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/physiology
14.
Cell ; 130(3): 427-39, 2007 Aug 10.
Article En | MEDLINE | ID: mdl-17693254

The autosomal dominant mutation in the human alphaB-crystallin gene inducing a R120G amino acid exchange causes a multisystem, protein aggregation disease including cardiomyopathy. The pathogenesis of cardiomyopathy in this mutant (hR120GCryAB) is poorly understood. Here, we show that transgenic mice overexpressing cardiac-specific hR120GCryAB recapitulate the cardiomyopathy in humans and find that the mice are under reductive stress. The myopathic hearts show an increased recycling of oxidized glutathione (GSSG) to reduced glutathione (GSH), which is due to the augmented expression and enzymatic activities of glucose-6-phosphate dehydrogenase (G6PD), glutathione reductase, and glutathione peroxidase. The intercross of hR120GCryAB cardiomyopathic animals with mice with reduced G6PD levels rescues the progeny from cardiac hypertrophy and protein aggregation. These findings demonstrate that dysregulation of G6PD activity is necessary and sufficient for maladaptive reductive stress and suggest a novel therapeutic target for abrogating R120GCryAB cardiomyopathy and heart failure in humans.


Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Mutation, Missense , Oxidative Stress/genetics , alpha-Crystallin B Chain/genetics , Animals , Arginine/genetics , Cardiomyopathies/enzymology , Glucosephosphate Dehydrogenase/antagonists & inhibitors , Glucosephosphate Dehydrogenase/genetics , Glucosephosphate Dehydrogenase/metabolism , Glutathione/metabolism , Glycine/genetics , Humans , Mice , Mice, Transgenic , Oxidation-Reduction , Proteins/metabolism , alpha-Crystallin B Chain/physiology
15.
J Chromatogr A ; 1130(2): 190-4, 2006 Oct 20.
Article En | MEDLINE | ID: mdl-16782116

The characterization of protein expression from a single-cell mouse embryo using two-dimensional capillary electrophoresis (2D-CE) is described. These zygotes were obtained from Hsf1 gene knockout mice. Single zygotes were lysed off-column and proteins were fluorescently labeled using the fluorogenic dye 3-(2-furoyl)quinoline-2-carboxaldehyde (FQ). After injection, analytes were separated first according to molecular weight using capillary sieving electrophoresis (CSE) and then by micellar electrokinetic capillary chromatography (MEKC) to obtain protein expression fingerprints. Analytes were detected in a sheath flow cuvette using laser-induced fluorescence. In a 1-h 2D-CE separation, over 100 components were resolved with a spot capacity of 380.


Electrophoresis, Capillary/methods , Electrophoresis, Gel, Two-Dimensional/methods , Embryo, Mammalian/chemistry , Proteins/analysis , Animals , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Mice , Mice, Knockout , Reproducibility of Results , Zygote/chemistry , Zygote/metabolism
16.
Methods ; 35(2): 170-5, 2005 Feb.
Article En | MEDLINE | ID: mdl-15649844

Since its discovery, stress or heat shock (HS) response has been widely studied as a paradigm for gene regulation. From control of gene expression to function and involvement in pathological processes, different aspects of the stress response have received extended attention and investigation by various approaches, using small analyzing molecules, cells and organisms. This chapter is focused on animal models, such as transgenic mice that allow integrated analysis of intact organisms in physiological and pathological conditions. Genetically modified mice, developed to generate gain- and loss-of-function, are described. The challenges of using the transgenic mouse model are also discussed.


Gene Expression Regulation , Heat-Shock Proteins/physiology , Mice, Transgenic , Animals , DNA/metabolism , Disease Models, Animal , Genes, Reporter , HSP70 Heat-Shock Proteins/metabolism , Hot Temperature , Humans , Mice , Mice, Knockout , Protein Conformation , Protein Folding , Temperature , Transgenes
17.
Med Sci (Paris) ; 19(4): 459-64, 2003 Apr.
Article Fr | MEDLINE | ID: mdl-12836219

Genes affected by maternal effect mutations encode maternal factors (transcripts, proteins) which are normally stored in oocytes and used by the embryos after fertilization. Although females bearing this type of mutation are viable and appear to be normal, embryonic development and survival of their offspring are compromised. Although maternal effect mutations are well known in lower organisms, such as drosophila or zebrafish, several examples have been only quite recently reported in mammals (Dnmt, Hsf1 and Mater). These studies provide new insights on the aspects of embryonic development directly controlled by maternal factors brought by the oocytes.


Gene Expression Regulation, Developmental , Mutation , Oocytes/physiology , Animals , Embryo, Nonmammalian/physiology , Embryonic and Fetal Development/genetics , Humans
18.
Semin Cell Dev Biol ; 14(5): 283-90, 2003 Oct.
Article En | MEDLINE | ID: mdl-14986858

Mammalian development follows a defined but adjustable program, depending on the plasticity of embryonic cells 'response to environmental changes. Heat shock proteins (Hsp) are integral part of this developmental program and gene targeting experiments have started to unravel developmental processes, which exhibit specific requirements for Hsps (e.g. Hsp70.2 for spermatogenesis). In the present paper, we will review available data on Hsp function and discuss the roles of heat shock factors (HSF), their major regulators, in mammalian development.


Embryonic and Fetal Development , Gene Expression Regulation, Developmental/physiology , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/metabolism , Spermatogenesis/physiology , Animals , Female , Humans , Male , Placenta/metabolism , Pregnancy
19.
EMBO J ; 21(19): 5164-72, 2002 Oct 01.
Article En | MEDLINE | ID: mdl-12356732

In this study, using heat shock factor 1 (Hsf1) knockout mice as a model, we tested the hypothesis that HSF1-dependent regulation of heat shock proteins (Hsps) is required to maintain redox state and attenuate oxidative damage in the normal heart. Here we report that, in mice, HSF1 deficiency reduces cardiac expression of Hsp25, alphaB-crystallin and Hsp70, but not Hsp60 and Hsp90. Consistent with the downregulation of Hsp25, for example, a significantly lower glutathione (GSH)/glutathione disulfate (GSSG) ratio was associated with the decreased activity, but not protein content, of glucose 6-phosphate dehydrogenase. Con sequently, superoxide was generated at a higher rate, and several mitochondrial proteins, including adenine nucleotide translocase 1 (ANT1), were more oxidized by HSF1 deficiency in vivo. Oxidative damage to ANT1 protein, a structural component of the mitochondrial permeability transition pore (MPTP), decreases its catalytic activity and increases MPTP opening, respectively. Taken together, our results indicate for the first time that constitutive expression of HSP chaperones requires HSF1 activity, and that such HSF1-dependent requirements are directly and functionally linked to maintain redox homeostasis and antioxidative defenses at normal (37 degrees C) temperature.


DNA-Binding Proteins/physiology , Gene Expression Regulation , Heart/physiology , Mitochondria, Heart/metabolism , Oxidative Stress/physiology , Animals , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Gene Expression Regulation/physiology , Glucosephosphate Dehydrogenase/metabolism , Glutathione/metabolism , Glutathione Disulfide/metabolism , Heat Shock Transcription Factors , Heat-Shock Proteins/deficiency , Heat-Shock Proteins/genetics , Heat-Shock Proteins/physiology , Homeostasis/genetics , Mice , Mice, Knockout , Oxidation-Reduction , Submitochondrial Particles/physiology , Superoxides/metabolism , Transcription Factors
20.
Crit Care Med ; 30(1 Supp): S43-S50, 2002 Jan.
Article En | MEDLINE | ID: mdl-11839944

OBJECTIVE: Life-threatening conditions cause severe changes in the organization and conformation of macromolecules, creating urgent requirements for protein repair to ensure survival. As molecular chaperones, heat shock proteins (HSP) that have specialized functions in protein folding are now well established to restore homeostasis in cells and organisms. Augmentation of HSP synthesis is tightly regulated by stress-inducible heat shock factors (HSF), which are part of a transcriptional signaling cascade with both positive (e.g., HSP) and negative (e.g., proinflammatory cytokines) properties. In this review, we discuss the biological roles and mechanisms of HSP-mediated protection in pathophysiologic conditions (ischemia, sepsis, and preeclampsia) and the regulation for stress-dependent HSP synthesis and speculate about future applications for harnessing HSF and HSP partners as cytoprotective agents. DATA SOURCES: Reactive oxygen species are major pathogenic factors in cell death pathways (e.g., necrosis, apoptosis), in part, because of proteotoxic effects. In intact organisms, forced overexpression of HSP per se affords effective counterbalance against ischemia challenges (e.g., heart and brain) and systemic conditions (e.g., sepsis). Besides stressful conditions, gene-targeting studies have uncovered new functions for heat shock transcription factors (e.g., maintenance of intrauterine pregnancy) in mammals. In parallel, pharmacologic studies using small molecules are paving the way for future prospects to exploit the beneficial properties of HSP, albeit an important but presently elusive goal. CONCLUSIONS: Together, HSF and HSP partners are attractive targets in therapeutic strategies designed to stimulate endogenous protective mechanisms against deleterious consequences of oxidative stress. With further technological advances, it is anticipated that the spotlight on HSP, alone or in combination with other stress response pathways, could, ultimately, reduce injury and accelerate functional recovery of susceptible organs in living organisms including humans.

...