Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Death Differ ; 27(7): 2280-2292, 2020 07.
Article in English | MEDLINE | ID: mdl-31996779

ABSTRACT

Heat shock can induce either cytoprotective mechanisms or cell death. We found that in certain human and mouse cells, including spermatocytes, activated heat shock factor 1 (HSF1) binds to sequences located in the intron(s) of the PMAIP1 (NOXA) gene and upregulates its expression which induces apoptosis. Such a mode of PMAIP1 activation is not dependent on p53. Therefore, HSF1 not only can activate the expression of genes encoding cytoprotective heat shock proteins, which prevents apoptosis, but it can also positively regulate the proapoptotic PMAIP1 gene, which facilitates cell death. This could be the primary cause of hyperthermia-induced elimination of heat-sensitive cells, yet other pro-death mechanisms might also be involved.


Subject(s)
Apoptosis , Heat Shock Transcription Factors/metabolism , Heat-Shock Response , Proto-Oncogene Proteins c-bcl-2/genetics , Signal Transduction , Up-Regulation/genetics , Animals , Apoptosis/genetics , Caspases/metabolism , Chromatin/metabolism , Enzyme Activation , Heat-Shock Response/genetics , Introns/genetics , Male , Mice, Knockout , Protein Binding , Proto-Oncogene Proteins c-bcl-2/deficiency , Tumor Suppressor Protein p53/metabolism
2.
Cancers (Basel) ; 11(10)2019 Oct 11.
Article in English | MEDLINE | ID: mdl-31614463

ABSTRACT

Heat Shock Factor 1 (HSF1) is a key regulator of gene expression during acute environmental stress that enables the cell survival, which is also involved in different cancer-related processes. A high level of HSF1 in estrogen receptor (ER)-positive breast cancer patients correlated with a worse prognosis. Here we demonstrated that 17ß-estradiol (E2), as well as xenoestrogen bisphenol A and ERα agonist propyl pyrazole triol, led to HSF1 phosphorylation on S326 in ERα positive but not in ERα-negative mammary breast cancer cells. Furthermore, we showed that MAPK signaling (via MEK1/2) but not mTOR signaling was involved in E2/ERα-dependent activation of HSF1. E2-activated HSF1 was transcriptionally potent and several genes essential for breast cancer cells growth and/or ERα action, including HSPB8, LHX4, PRKCE, WWC1, and GREB1, were activated by E2 in a HSF1-dependent manner. Our findings suggest a hypothetical positive feedback loop between E2/ERα and HSF1 signaling, which may support the growth of estrogen-dependent tumors.

3.
Int J Mol Sci ; 21(1)2019 Dec 30.
Article in English | MEDLINE | ID: mdl-31906015

ABSTRACT

Spermatocytes are among the most heat-sensitive cells and the exposure of testes to heat shock results in their Heat Shock Factor 1 (HSF1)-mediated apoptosis. Several lines of evidence suggest that pleckstrin-homology-like domain family A, member 1 (PHLDA1) plays a role in promoting heat shock-induced cell death in spermatogenic cells, yet its precise physiological role is not well understood. Aiming to elucidate the hypothetical role of PHLDA1 in HSF1-mediated apoptosis of spermatogenic cells we characterized its expression in mouse testes during normal development and after heat shock. We stated that transcription of Phlda1 is upregulated by heat shock in many adult mouse organs including the testes. Analyzes of the Phlda1 expression during postnatal development indicate that it is expressed in pre-meiotic or somatic cells of the testis. It starts to be transcribed much earlier than spermatocytes are fully developed and its transcripts and protein products do not accumulate further in the later stages. Moreover, neither heat shock nor expression of constitutively active HSF1 results in the accumulation of PHLDA1 protein in meiotic and post-meiotic cells although both conditions induce massive apoptosis of spermatocytes. Furthermore, the overexpression of PHLDA1 in NIH3T3 cells leads to cell detachment, yet classical apoptosis is not observed. Therefore, our findings indicate that PHLDA1 cannot directly contribute to the heat-induced apoptosis of spermatocytes. Instead, PHLDA1 could hypothetically participate in death of spermatocytes indirectly via activation of changes in the somatic or pre-meiotic cells present in the testes.


Subject(s)
Apoptosis/drug effects , Apoptosis/physiology , Heat Shock Transcription Factors/pharmacology , Spermatocytes/metabolism , Transcription Factors/metabolism , Animals , Animals, Genetically Modified , Cloning, Molecular , Heat-Shock Response/physiology , Male , Mice , NIH 3T3 Cells , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Testis/metabolism , Testis/pathology , Transcription Factors/genetics
4.
Reproduction ; 156(3): 195-206, 2018 09.
Article in English | MEDLINE | ID: mdl-29880719

ABSTRACT

SPEN (spen family transcription repressor) is a nucleic acid-binding protein putatively involved in repression of gene expression. We hypothesized that SPEN could be involved in general downregulation of the transcription during the heat shock response in mouse spermatogenic cells through its interactions with chromatin. We documented predominant nuclear localization of the SPEN protein in spermatocytes and round spermatids, which was retained after heat shock. Moreover, the protein was excluded from the highly condensed chromatin. Chromatin immunoprecipitation experiments clearly indicated interactions of SPEN with chromatin in vivo However, ChIP-Seq analyses did not reveal any strong specific peaks both in untreated and heat shocked cells, which might suggest dispersed localization of SPEN and/or its indirect binding to DNA. Using in situ proximity ligation assay we found close in vivo associations of SPEN with MTA1 (metastasis-associated 1), a member of the nucleosome remodeling complex with histone deacetylase activity, which might contribute to interactions of SPEN with chromatin.


Subject(s)
Chromatin/metabolism , Gene Expression Regulation/physiology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Animals , Cell Nucleus/chemistry , Chromatin/chemistry , DNA-Binding Proteins , Histone Deacetylases/metabolism , Hot Temperature , Male , Mice , Mice, Inbred Strains , Nuclear Proteins/analysis , RNA-Binding Proteins , Repressor Proteins , Spermatids/ultrastructure , Spermatocytes/ultrastructure , Spermatogenesis , Testis/cytology , Trans-Activators , Transcription Factors/metabolism
5.
PLoS One ; 10(12): e0143688, 2015.
Article in English | MEDLINE | ID: mdl-26625260

ABSTRACT

BACKGROUND: The molecular mechanisms driving the papillary thyroid carcinoma (PTC) are still poorly understood. The most frequent genetic alteration in PTC is the BRAFV600E mutation--its impact may extend even beyond PTC genomic profile and influence the tumor characteristics and even clinical behavior. METHODS: In order to identify BRAF-dependent signature of early carcinogenesis in PTC, a transgenic mouse model with BRAFV600E-induced PTC was developed. Mice thyroid samples were used in microarray analysis and the data were referred to a human thyroid dataset. RESULTS: Most of BRAF(+) mice developed malignant lesions. Nevertheless, 16% of BRAF(+) mice displayed only benign hyperplastic lesions or apparently asymptomatic thyroids. After comparison of non-malignant BRAF(+) thyroids to BRAF(-) ones, we selected 862 significantly deregulated genes. When the mouse BRAF-dependent signature was transposed to the human HG-U133A microarray, we identified 532 genes, potentially indicating the BRAF signature (representing early changes, not related to developed malignant tumor). Comparing BRAF(+) PTCs to healthy human thyroids, PTCs without BRAF and RET alterations and RET(+), RAS(+) PTCs, 18 of these 532 genes displayed significantly deregulated expression in all subgroups. All 18 genes, among them 7 novel and previously not reported, were validated as BRAFV600E-specific in the dataset of independent PTC samples, made available by The Cancer Genome Atlas Project. CONCLUSION: The study identified 7 BRAF-induced genes that are specific for BRAF V600E-driven PTC and not previously reported as related to BRAF mutation or thyroid carcinoma: MMD, ITPR3, AACS, LAD1, PVRL3, ALDH3B1, and RASA1. The full signature of BRAF-related 532 genes may encompass other BRAF-related important transcripts and require further study.


Subject(s)
Carcinoma/genetics , Gene Expression Regulation, Neoplastic , Mutation, Missense , Proto-Oncogene Proteins B-raf/genetics , Thyroid Neoplasms/genetics , Transcriptome , Animals , Carcinoma/metabolism , Carcinoma, Papillary , Female , Humans , Male , Mice , Mice, Transgenic , Microarray Analysis , Thyroid Cancer, Papillary , Thyroid Neoplasms/metabolism
6.
Cell Signal ; 27(2): 394-401, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25435429

ABSTRACT

Heat shock transcription factor 1 (HSF1), the major regulator of stress response, is frequently activated in cancer and has an apparent role in malignant transformation. Here we analyzed the influence of the over-expression of a constitutively active transcriptionally-competent HSF1 mutant form on phenotypes of mouse and human melanoma cells. We observed that the expression of active HSF1 supported anchorage-independent growth in vitro, and metastatic spread in the animal model in vivo, although the proliferation rate of cancer cells was not affected. Furthermore, active HSF1 enhanced cell motility, reduced the adherence of cells to a fibronectin-coated surface, and affected the actin cytoskeleton. We found that although the expression of active HSF1 did not affect levels of epithelial-to-mesenchymal transition markers, it caused transcriptional down-regulation of vinculin, protein involved in cell motility, and adherence. Functional HSF1-binding sites were found in mouse and human Vcl/VCL genes, indicating a direct role of HSF1 in the regulation of this gene. An apparent association between HSF1-induced down-regulation of vinculin, increased motility, and a reduced adherence of cells suggests a possible mechanism of HSF1-mediated enhancement of the metastatic potential of cancer cells.


Subject(s)
DNA-Binding Proteins/metabolism , Melanoma/pathology , Transcription Factors/metabolism , Vinculin/metabolism , Actin Cytoskeleton/metabolism , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Down-Regulation , Epithelial-Mesenchymal Transition , Female , Heat Shock Transcription Factors , Humans , Melanoma/metabolism , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Temperature , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transplantation, Homologous , Vinculin/genetics
7.
Int J Biochem Cell Biol ; 57: 76-83, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25450459

ABSTRACT

Heat Shock Factor 1 (HSF1) is the primary transcription factor responsible for the response to cellular stress, while HSF2 becomes activated during development and differentiation, including spermatogenesis. Although both factors are indispensable for proper spermatogenesis, activation of HSF1 by heat shock initiates apoptosis of spermatogenic cells leading to infertility of males. To characterize mechanisms assisting such heat induced apoptosis we studied how HSF1 and HSF2 cooperate during the heat shock response. For this purpose we used chromatin immunoprecipitation and the proximity ligation approaches. We looked for co-occupation of binding sites by HSF1 and HSF2 in untreated (32 °C) or heat shocked (at 38 °C or 43 °C) spermatocytes, which are cells the most sensitive to hyperthermia. At the physiological temperature or after mild hyperthermia at 38 °C, the sharing of binding sites for both HSFs was observed mainly in promoters of Hsp genes and other stress-related genes. Strong hyperthermia at 43 °C resulted in an increased binding of HSF1 and releasing of HSF2, hence co-occupation of promoter regions was not detected any more. The close proximity of HSF1 and HSF2 (and/or existence of HSF1/HSF2 complexes) was frequent at the physiological temperature. Temperature elevation resulted in a decreased number of such complexes and they were barely detected after strong hyperthermia at 43 °C. We have concluded that at the physiological temperature HSF1 and HSF2 cooperate in spermatogenic cells. However, temperature elevation causes remodeling of chromatin binding and interactions between HSFs are disrupted. This potentially affects the regulation of stress response and contributes to the heat sensitivity of these cells.


Subject(s)
DNA-Binding Proteins/metabolism , Heat-Shock Proteins/metabolism , Heat-Shock Response/physiology , Testis/metabolism , Transcription Factors/metabolism , Animals , Binding Sites , Chromatin/metabolism , Chromatin Immunoprecipitation , DNA-Binding Proteins/genetics , Genome-Wide Association Study , Heat Shock Transcription Factors , Heat-Shock Proteins/genetics , Heat-Shock Response/genetics , Hyperthermia, Induced , Male , Mice , Promoter Regions, Genetic , Spermatocytes/metabolism , Testis/cytology , Transcription Factors/genetics
8.
BMC Genomics ; 14: 456, 2013 Jul 08.
Article in English | MEDLINE | ID: mdl-23834426

ABSTRACT

BACKGROUND: Elevated temperatures induce activation of the heat shock transcription factor 1 (HSF1) which in somatic cells leads to heat shock proteins synthesis and cytoprotection. However, in the male germ cells (spermatocytes) caspase-3 dependent apoptosis is induced upon HSF1 activation and spermatogenic cells are actively eliminated. RESULTS: To elucidate a mechanism of such diverse HSF1 activity we carried out genome-wide transcriptional analysis in control and heat-shocked cells, either spermatocytes or hepatocytes. Additionally, to identify direct molecular targets of active HSF1 we used chromatin immunoprecipitation assay (ChIP) combined with promoter microarrays (ChIP on chip). Genes that are differently regulated after HSF1 binding during hyperthermia in both types of cells have been identified. Despite HSF1 binding to promoter sequences in both types of cells, strong up-regulation of Hsps and other genes typically activated by the heat shock was observed only in hepatocytes. In spermatocytes HSF1 binding correlates with transcriptional repression on a large scale. HSF1-bound and negatively regulated genes encode mainly for proteins required for cell division, involved in RNA processing and piRNA biogenesis. CONCLUSIONS: Observed suppression of the transcription could lead to genomic instability caused by meiotic recombination disturbances, which in turn might induce apoptosis of spermatogenic cells. We propose that HSF1-dependent induction of cell death is caused by the simultaneous repression of many genes required for spermatogenesis, which guarantees the elimination of cells damaged during heat shock. Such activity of HSF1 prevents transmission of damaged genetic material to the next generation.


Subject(s)
Apoptosis/genetics , DNA-Binding Proteins/metabolism , Gene Expression Profiling , Heat-Shock Response/genetics , Hepatocytes/cytology , Spermatocytes/cytology , Transcription Factors/metabolism , Animals , Chromatin Immunoprecipitation , DNA/metabolism , Heat Shock Transcription Factors , Hepatocytes/metabolism , Male , Mice , Oligonucleotide Array Sequence Analysis , Spermatocytes/metabolism , Spermatogenesis/genetics , Transcription, Genetic
9.
Reproduction ; 143(6): 749-57, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22495889

ABSTRACT

The binding of capacitated spermatozoa to the egg's extracellular coat and induction of acrosome reaction are necessary for successful fertilization in mammals. Biogenesis of acrosome is complicated, and not all proteins involved in this process are known. In this study, we have cloned a novel mouse gene, Spaca7, that is expressed exclusively in the testes. During the postnatal development, transcripts of the gene could be detected at a very low level in 18-day-old mouse testes and at a higher level in 21-day-old mouse testes and later, which corresponds to an expansion of round spermatids. In the stably transfected PT67 cells, SPACA7 fused with EGFP was predominantly localized in the Golgi apparatus. In transgenic mouse testes, the fusion protein was found in acrosome (starting from the first stages of acrosome formation in late pachytene spermatocytes and finally in spermatozoa isolated from caput and cauda of epididymis). Confocal microscopy studies revealed an intra-acrosomal not membrane-bound localization of SPACA7/EGFP, which suggests that the protein can be released during acrosome reaction and involved in fertilization. Acrosomal localization of endogenous SPACA7 protein was also found in human spermatozoa.


Subject(s)
Acrosome Reaction/genetics , Seminal Plasma Proteins/isolation & purification , Animals , Cells, Cultured , Cloning, Molecular , Fertilization/genetics , Fertilization/physiology , Humans , Male , Membrane Proteins/genetics , Membrane Proteins/isolation & purification , Membrane Proteins/metabolism , Mice , Mice, Transgenic , NIH 3T3 Cells , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Seminal Plasma Proteins/genetics , Seminal Plasma Proteins/metabolism , Spermatogenesis/genetics , Spermatogenesis/physiology , Spermatozoa/chemistry , Spermatozoa/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...