Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cells ; 10(12)2021 12 07.
Article in English | MEDLINE | ID: mdl-34943954

ABSTRACT

The high frequency of breast cancer worldwide and the high mortality among women with this malignancy are a serious challenge for modern medicine. A deeper understanding of the mechanisms of carcinogenesis and emergence of metastatic, therapy-resistant breast cancers would help development of novel approaches to better treatment of this disease. The review is dedicated to the role of members of the heat shock protein 70 subfamily (HSP70s or HSPA), mainly inducible HSP70, glucose-regulated protein 78 (GRP78 or HSPA5) and GRP75 (HSPA9 or mortalin), in the development and pathogenesis of breast cancer. Various HSP70-mediated cellular mechanisms and pathways which contribute to the oncogenic transformation of mammary gland epithelium are reviewed, as well as their role in the development of human breast carcinomas with invasive, metastatic traits along with the resistance to host immunity and conventional therapeutics. Additionally, intracellular and cell surface HSP70s are considered as potential targets for therapy or sensitization of breast cancer. We also discuss a clinical implication of Hsp70s and approaches to targeting breast cancer with gene vectors or nanoparticles downregulating HSP70s, natural or synthetic (small molecule) inhibitors of HSP70s, HSP70-binding antibodies, HSP70-derived peptides, and HSP70-based vaccines.


Subject(s)
Breast Neoplasms/genetics , Carcinogenesis/genetics , Endoplasmic Reticulum Chaperone BiP/genetics , HSP70 Heat-Shock Proteins/genetics , Mitochondrial Proteins/genetics , Breast Neoplasms/pathology , Female , Humans , Membrane Proteins/genetics
2.
Cancers (Basel) ; 13(5)2021 Mar 04.
Article in English | MEDLINE | ID: mdl-33806538

ABSTRACT

Within aggressive malignancies, there usually are the "hypoxic zones"-poorly vascularized regions where tumor cells undergo oxygen deficiency through inadequate blood supply. Besides, hypoxia may arise in tumors as a result of antiangiogenic therapy or transarterial embolization. Adapting to hypoxia, tumor cells acquire a hypoxia-resistant phenotype with the characteristic alterations in signaling, gene expression and metabolism. Both the lack of oxygen by itself and the hypoxia-responsive phenotypic modulations render tumor cells more radioresistant, so that hypoxic tumors are a serious challenge for radiotherapy. An understanding of causes of the radioresistance of hypoxic tumors would help to develop novel ways for overcoming this challenge. Molecular targets for and various approaches to radiosensitizing hypoxic tumors are considered in the present review. It is here analyzed how the hypoxia-induced cellular responses involving hypoxia-inducible factor-1, heat shock transcription factor 1, heat shock proteins, glucose-regulated proteins, epigenetic regulators, autophagy, energy metabolism reprogramming, epithelial-mesenchymal transition and exosome generation contribute to the radioresistance of hypoxic tumors or may be inhibited for attenuating this radioresistance. The pretreatments with a multitarget inhibition of the cancer cell adaptation to hypoxia seem to be a promising approach to sensitizing hypoxic carcinomas, gliomas, lymphomas, sarcomas to radiotherapy and, also, liver tumors to radioembolization.

3.
Cells ; 9(4)2020 04 06.
Article in English | MEDLINE | ID: mdl-32268506

ABSTRACT

Cancer stem cells (CSCs) are a great challenge in the fight against cancer because these self-renewing tumorigenic cell fractions are thought to be responsible for metastasis dissemination and cases of tumor recurrence. In comparison with non-stem cancer cells, CSCs are known to be more resistant to chemotherapy, radiotherapy, and immunotherapy. Elucidation of mechanisms and factors that promote the emergence and existence of CSCs and their high resistance to cytotoxic treatments would help to develop effective CSC-targeting therapeutics. The present review is dedicated to the implication of molecular chaperones (protein regulators of polypeptide chain folding) in both the formation/maintenance of the CSC phenotype and cytoprotective machinery allowing CSCs to survive after drug or radiation exposure and evade immune attack. The major cellular chaperones, namely heat shock proteins (HSP90, HSP70, HSP40, HSP27), glucose-regulated proteins (GRP94, GRP78, GRP75), tumor necrosis factor receptor-associated protein 1 (TRAP1), peptidyl-prolyl isomerases, protein disulfide isomerases, calreticulin, and also a transcription heat shock factor 1 (HSF1) initiating HSP gene expression are here considered as determinants of the cancer cell stemness and potential targets for a therapeutic attack on CSCs. Various approaches and agents are discussed that may be used for inhibiting the chaperone-dependent development/manifestations of cancer cell stemness.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Neoplastic Stem Cells/metabolism , Cell Line, Tumor , Endoplasmic Reticulum Chaperone BiP , Humans
4.
Methods Mol Biol ; 1709: 107-127, 2018.
Article in English | MEDLINE | ID: mdl-29177655

ABSTRACT

Heat shock proteins are well-known protectors from cell death. Cell death (in particular, apoptosis and necrosis) is accompanied by certain hallmarks manifested as specific alterations in cellular membranes, cytoplasm, nucleus, and mitochondria. Some of those hallmarks are easily detectable in situ and, therefore, they can be applied for the assessment of dying or dead cells. In turn, there are also signs of viable cells that include such features as normal functioning of their membranes and organelles, ability to proliferate, etc. This chapter describes several convenient methods for quantification of dead (apoptotic and necrotic) cells as well as methods for assessment of viable cells. We describe in detail methods of annexin V/propidium iodide (PI) staining, TUNEL assay, Hoechst/PI staining, caspase activation, MTS tetrazolium, lactate dehydrogenase (LDH) release, colony formation, and senescence assays, with the principles, advantages, and drawbacks of each technique.


Subject(s)
Apoptosis , Biological Assay/methods , Flow Cytometry/methods , In Situ Nick-End Labeling/methods , Necrosis , Animals , Annexin A5 , Fluorescent Dyes , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/physiology , Humans , L-Lactate Dehydrogenase , Propidium , Tetrazolium Salts
5.
PLoS One ; 12(3): e0173640, 2017.
Article in English | MEDLINE | ID: mdl-28291803

ABSTRACT

We studied a role of the inducible heat shock protein 70 (Hsp70) in cellular response to radiosensitizing treatments with inhibitors of the heat shock protein 90 (Hsp90) chaperone activity. Cell lines derived from solid tumors of different origin were treated with the Hsp90 inhibitors (17AAG, geldanamycin, radicicol, NVP-AUY922) or/and γ-photon radiation. For comparison, human cells of the non-cancerous origin were subjected to the same treatments. We found that the Hsp90 inhibitors yielded considerable radiosensitization only when they cause early and pronounced Hsp70 induction; moreover, a magnitude of radiosensitization was positively correlated with the level of Hsp70 induction. The quantification of Hsp70 levels in Hsp90 inhibitor-treated normal and cancer cells enabled to predict which of them will be susceptible to any Hsp90-inhibiting radiosensitizer as well as what concentrations of the inhibitors ensure the preferential cytotoxicity in the irradiated tumors without aggravating radiation damage to adjacent normal tissues. Importantly, the Hsp70 induction in the Hsp90 inhibitor-treated cancer cells appears to be their protective response that alleviates the tumor-sensitizing effects of the Hsp90 inactivation. Combination of the Hsp70-inducing inhibitors of Hsp90 with known inhibitors of the Hsp induction such as quercetin, triptolide, KNK437, NZ28 prevented up-regulation of Hsp70 in the cancer cells thereby increasing their post-radiation apoptotic/necrotic death and decreasing their post-radiation viability/clonogenicity. Similarly, co-treatment with the two inhibitors conferred the enhanced radiosensitization of proliferating rather than quiescent human vascular endothelial cells which may be used for suppressing the tumor-stimulated angiogenesis. Thus, the easily immunodetectable Hsp70 induction can be a useful marker for predicting effects of Hsp90-inhibiting radiosensitizers on tumors and normal tissues exposed to ionizing radiation. Moreover, targeting the Hsp70 induction in Hsp90 inhibitor-treated cancer cells and tumor vasculature cells may beneficially enhance the radiosensitizing effect.


Subject(s)
Biomarkers/metabolism , HSP70 Heat-Shock Proteins/biosynthesis , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Radiation-Sensitizing Agents/pharmacology , Cell Line, Tumor , Humans
6.
Methods Mol Biol ; 787: 231-44, 2011.
Article in English | MEDLINE | ID: mdl-21898239

ABSTRACT

Cell death (in particular, apoptosis and necrosis) is accompanied by appearance of certain hallmarks that are manifested as specific alterations in cellular membranes, cytoplasm, nucleus and mitochondria. Some of those hallmarks are easily detectable in situ and, therefore, they can be applied for the assessment of dying or dead cells. In turn, there are also signs of viable cells that include a set of features, such as normal functioning of their membranes and organelles, ability to proliferate, etc. The present chapter provides descriptions of several convenient methods for quantitative determination of dead (apoptotic and necrotic) cells and also methods for determination of survived and viable cells. Here, we describe in details the methods of annexin V/propidium iodide (PI) staining, TUNEL assay, Hoechst/PI staining, MTS tetrazolium assay, and colony formation assay, with the principles, advantages, and drawbacks of each technique.


Subject(s)
Cell Death/physiology , Cell Survival/physiology , Heat-Shock Proteins/analysis , Annexin A5/analysis , Cell Membrane , Humans , In Situ Nick-End Labeling/methods , Propidium , Staining and Labeling/methods , Tetrazolium Salts , Tumor Stem Cell Assay/methods
7.
J Mol Med (Berl) ; 88(3): 241-7, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19946660

ABSTRACT

The 90-kD heat shock protein (Hsp90) is an abundant molecular chaperone catalyzing maturation and activation of client proteins. A number of the Hsp90 client proteins are components of cancer cell-associated signaling pathways that ensure unlimited growth of tumors and their resistance to chemotherapy and radiotherapy. Upon inhibition of the Hsp90 chaperone function, such client proteins are destabilized and degraded which disrupts multiple pathways essential for tumor cell survival; hence, pharmacological Hsp90 inhibitors could be applied in anticancer therapy. Several Hsp90-inhibiting compounds are currently tested in preclinical or phase I-III clinical trials as single anticancer agents or in combination with conventional drugs and radiation. The present review summarizes the data characterizing Hsp90 inhibitors as agents that sensitize human tumors to irradiation which may improve the outcome of radiotherapy. We also discuss molecular mechanisms of the Hsp90 inhibition-induced radiosensitization and its selectivity toward cancer cells.


Subject(s)
HSP90 Heat-Shock Proteins/antagonists & inhibitors , Neoplasms/radiotherapy , Radiation-Sensitizing Agents , Animals , Antineoplastic Agents/therapeutic use , Clinical Trials, Phase I as Topic , Combined Modality Therapy , HSP90 Heat-Shock Proteins/metabolism , Humans , Neoplasms/drug therapy , Radiation-Sensitizing Agents/chemistry
8.
Int J Radiat Oncol Biol Phys ; 71(3): 858-65, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18410996

ABSTRACT

PURPOSE: In addition to invasive tumor cells, endothelial cells (ECs) of the tumor vasculature are an important target for anticancer radiotherapy. The purpose of the present work is to investigate how 17-N-allilamino-17-demethoxygeldanamycin (17AAG), known as an anticancer drug inhibiting heat shock protein 90 (Hsp90), modifies radiation responses of human vascular ECs. METHODS AND MATERIALS: The ECs cultured from human umbilical veins were exposed to gamma-irradiation, whereas some EC samples were pretreated with growth factors and/or 17AAG. Postirradiation cell death/survival and morphogenesis were assessed by means of terminal deoxynucleotidyl transferase biotin-deoxyuridine triphosphate nick end labeling or annexin V staining and clonogenic and tube-formation assays. The 17AAG-affected expression and phosphorylation of radioresistance-related proteins were probed by means of immunoblotting. Dominant negative or constitutively activated Akt was transiently expressed in ECs to manipulate Akt activity. RESULTS: It was found that nanomolar concentrations of 17AAG sensitize ECs to relatively low doses (2-6 Gy) of gamma-irradiation and abolish the radioprotective effects of vascular endothelial growth factor and basic fibroblast growth factor. The drug-induced radiosensitization of ECs seems to be caused by prevention of Hsp90-dependent phosphorylation (activation) of Akt that results in blocking the radioprotective phosphatidylinositol 3-kinase/Akt pathway. CONCLUSIONS: Clinically achievable concentrations of 17AAG can decrease the radioresistance intrinsic to vascular ECs and minimize the radioprotection conferred upon them by tumor-derived growth factors. These findings characterize 17AAG as a promising radiosensitizer for the tumor vasculature.


Subject(s)
Benzoquinones/administration & dosage , Endothelial Cells/physiology , Endothelial Cells/radiation effects , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Lactams, Macrocyclic/administration & dosage , Radiation Tolerance/physiology , Cells, Cultured , Endothelial Cells/drug effects , Humans , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/administration & dosage
9.
Biochem J ; 407(3): 407-17, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17650072

ABSTRACT

In vitro, small Hsps (heat-shock proteins) have been shown to have chaperone function capable of keeping unfolded proteins in a form competent for Hsp70-dependent refolding. However, this has never been confirmed in living mammalian cells. In the present study, we show that Hsp27 (HspB1) translocates into the nucleus upon heat shock, where it forms granules that co-localize with IGCs (interchromatin granule clusters). Although heat-induced changes in the oligomerization status of Hsp27 correlate with its phosphorylation and nuclear translocation, Hsp27 phosphorylation alone is not sufficient for effective nuclear translocation of HspB1. Using firefly luciferase as a heat-sensitive reporter protein, we demonstrate that HspB1 expression in HspB1-deficient fibroblasts enhances protein refolding after heat shock. The positive effect of HspB1 on refolding is completely diminished by overexpression of Bag-1 (Bcl-2-associated athanogene), the negative regulator of Hsp70, consistent with the idea of HspB1 being the substrate holder for Hsp70. Although HspB1 and luciferase both accumulate in nuclear granules after heat shock, our results suggest that this is not related to the refolding activity of HspB1. Rather, granular accumulation may reflect a situation of failed refolding where the substrate is stored for subsequent degradation. Consistently, we found 20S proteasomes concentrated in nuclear granules of HspB1 after heat shock. We conclude that HspB1 contributes to an increased chaperone capacity of cells by binding unfolded proteins that are hereby kept competent for refolding by Hsp70 or that are sorted to nuclear granules if such refolding fails.


Subject(s)
Heat-Shock Proteins/physiology , Heat-Shock Response/physiology , Intracellular Fluid/physiology , Molecular Chaperones/physiology , Neoplasm Proteins/physiology , Animals , Cell Line , Cell Nucleus Structures/chemistry , Cell Nucleus Structures/metabolism , HSP27 Heat-Shock Proteins , HSP70 Heat-Shock Proteins/physiology , Intracellular Fluid/chemistry , Mice , Oxidative Stress/physiology , Protein Folding , Protein Sorting Signals/physiology , Protein Transport/physiology , Rats
10.
Radiat Res ; 165(4): 410-23, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16579653

ABSTRACT

To elucidate how the heat-shock transcription factor 1 (Hsf1)-mediated stress response affects cellular radioresistance, mouse embryo fibroblasts with Hsf1-gene knockout (Hsf1(-/-) cells) or with normal wild-type Hsf1 expression (Hsf1 wild-type cells) were preconditioned by heating (43 degrees C, 30 min) without or with quercetin (an inhibitor of Hsf1) and then exposed to gamma radiation (4 or 6 Gy). Some cell samples were infected with virus-based vectors to overexpress the constitutively active (mutant) form of Hsf1 or individual heat-shock proteins (Hsps). The heat preconditioning transiently up-regulated the Hsp levels in Hsf1 wild-type cells and significantly improved their postirradiation survival; these effects could be abolished by quercetin or simulated (without preheating) by the Hsf1 overexpression. In contrast, no enhanced radioresistance was found in heat-preconditioned Hsf1(-/-) cells that were unable to trigger Hsf1-mediated Hsp induction after heating. However, when the constitutively active Hsf1 was overexpressed in Hsf1(-/-) cells, the latter accumulated stress-inducible Hsps and became more radioresistant like heat-preconditioned Hsf1 wild-type cells. The overexpression of Hsp70 or/ and Hsp27 also enhanced radioresistance of both cell cultures. Thus the preirradiation stress response resulting in the intracellular Hsp accumulation can improve survival of severely irradiated mammalian cells.


Subject(s)
Apoptosis/physiology , Apoptosis/radiation effects , DNA-Binding Proteins/metabolism , Fibroblasts/physiology , Fibroblasts/radiation effects , Heat-Shock Response/physiology , Heat-Shock Response/radiation effects , Transcription Factors/metabolism , Animals , Cells, Cultured , Dose-Response Relationship, Radiation , Fibroblasts/cytology , Heat Shock Transcription Factors , Mice , Radiation Dosage , Radiation Tolerance/physiology
11.
Mol Cell Biol ; 23(14): 4948-58, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12832480

ABSTRACT

CHIP is a cochaperone of Hsp70 that inhibits Hsp70-dependent refolding in vitro. However, the effect of altered expression of CHIP on the fate of unfolded proteins in mammalian cells has not been determined. Surprisingly, we found that overexpression of CHIP in fibroblasts increased the refolding of proteins after thermal denaturation. This effect was insensitive to geldanamycin, an Hsp90 inhibitor, and required the tetratricopeptide repeat motifs but not the U-box domain of CHIP. Inhibition of Hsp70 chaperone activity abolished the effects of CHIP on protein folding, indicating that the CHIP-mediated events were Hsp70 dependent. Hsp40 competitively inhibited the CHIP-dependent refolding, which is consistent with in vitro data indicating that these cofactors act on Hsp70 in the ATP-bound state and have opposing effects on Hsp70 ATPase activity. Consistent with these observations, CHIP overexpression did not alter protein folding in the setting of ATP depletion, when Hsp70 is in the ADP-bound state. Concomitant with its effects on refolding heat-denatured substrates, CHIP increased the fraction of nascent chains coimmunoprecipitating with Hsc70, but only when sufficient ATP was present to allow Hsp70 to cycle rapidly. Our data suggest that, consistent with in vitro studies, CHIP attenuates the Hsp70 cycle in living cells. The impact of this effect on the fate of unfolded proteins in cells, however, is different from what might be expected from the in vitro data. Rather than resulting in inhibited refolding, CHIP increases the folding capacity of Hsp70 in eukaryotic cells.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Nuclear Proteins/metabolism , Protein Folding , Adenosine Triphosphate/metabolism , Animals , Benzoquinones , CHO Cells , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cricetinae , DNA-Binding Proteins , Enzyme Activation , HSP40 Heat-Shock Proteins , HSP70 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Lactams, Macrocyclic , Luciferases/genetics , Luciferases/metabolism , Mammals , Mutation , Nuclear Proteins/genetics , Proteins/metabolism , Quinones/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repetitive Sequences, Amino Acid , Transcription Factors
12.
Cell Stress Chaperones ; 8(4): 335-47, 2003.
Article in English | MEDLINE | ID: mdl-15115286

ABSTRACT

Overexpression of heat shock protein (Hsp) 70 and Hsp27 in vivo was proclaimed as a potential tool in therapy of ischemia-reperfusion injury. However, it was so far not known whether these Hsps can beneficially act when increased in cells just at the stage of postischemic reperfusion. This issue was examined in a model of ischemia-reperfusion stress when cultures of endothelial cells (EC) from human umbilical vein were infected with virus-based vectors expressing Hsp70 or Hsp27, or Hsp56, or green fluorescent protein (GFP) and exposed to 20 hours of hypoxia followed by reoxygenation. The infection was performed either 10 hours before hypoxia or immediately after hypoxia, or at different time points of reoxygenation. Only low cell death was detected during hypoxia, but later, up to 40% of the treated cells died via caspase-dependent apoptosis between 6 and 12 hours of reoxygenation. The percentage of apoptotic cells was 1.6- to 3-fold greater in Hsp56- and GFP-infected EC than in Hsp70- or Hsp27-infected EC. The last 2 groups exhibited a lesser extent of procaspase-9 and procaspase-3 activation within 6-9 hours of reoxygenation. The cytoprotective effects of overexpressed Hsp70 and Hsp27 were observed not only in the case of infection before hypoxia but also when EC were infected at the start of reoxygenation or 1-2 hours later. An increase in the Hsp70 and Hsp27 levels in infected EC correlated well with their resistance to apoptosis under reoxygenation. These findings suggest that overexpression of Hsp70 or Hsp27, if it occurs in the involved cells at the early stage of postischemic reperfusion, can still be cytoprotective.


Subject(s)
Apoptosis/physiology , Endothelial Cells/metabolism , HSP70 Heat-Shock Proteins/genetics , Reperfusion Injury/therapy , Caspases/metabolism , Genes, Reporter , HSP70 Heat-Shock Proteins/biosynthesis , Humans , Hypoxia/metabolism , Reperfusion , Reperfusion Injury/metabolism , Time Factors , Umbilical Veins/metabolism
13.
Cell Stress Chaperones ; 7(2): 146-55, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12380682

ABSTRACT

The behavior of the endogenous heat shock protein 25 (Hsp25) in heat-stressed rat H9c2 myoblasts was studied. After mild or severe heating, this protein became less extractable with Triton X-100 and displayed characteristic immunofluorescence patterns, namely (1) granules in the nucleus, and (2) association with F-actin bundles in the cytoplasm. The intranuclear granulation of Hsp25 and its association with F-actin were sensitive to drugs affecting Hsp25 phosphorylation (cantharidin, sodium orthovanadate, SB203580, SB202190). Isoform analysis of Hsp25 translocated to the nucleus-free cytoskeletal fraction revealed only mono- and biphosphorylated Hsp25 and no unphosphorylated Hsp25. Transfected luciferase with initial localization in the nucleosol became colocalized with the Hsp25-containing granules after a heat shock treatment that denatured the enzyme in the cells. The association of Hsp25 with actin filaments after a mild heat stress conferred protection from subsequent F-actin-damaging treatments with cytochalasins (D and B) or severe heat stress. We hypothesize that (1) the binding of heat-denatured nucleosolic proteins to the Hsp25 contained in specific granular structures may serve for the subsequent chaperoning or degradation of the bound proteins, and (2) the actin cytoskeleton is stabilized by the direct targeting of phosphorylated Hsp25 to microfilament bundles.


Subject(s)
Heat-Shock Proteins , Hot Temperature , Myoblasts/metabolism , Neoplasm Proteins/metabolism , Animals , Cell Nucleus/metabolism , Cell Survival , Cytoskeleton/metabolism , HSP27 Heat-Shock Proteins , Imidazoles/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Pyridines/metabolism , Rats , p38 Mitogen-Activated Protein Kinases
14.
Am J Physiol Cell Physiol ; 283(2): C521-34, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12107062

ABSTRACT

Rat H9c2 myoblasts were preconditioned by heat or metabolic stress followed by recovery under normal conditions. Cells were then subjected to severe ATP depletion, and stress-associated proteotoxicity was assessed on 1) the increase in a Triton X-100-insoluble component of total cellular protein and 2) the rate of inactivation and insolubilization of transfected luciferase with cytoplasmic or nuclear localization. Both heat and metabolic preconditioning elevated the intracellular heat shock protein 70 (HSP70) level and reduced cell death after sustained ATP depletion without affecting the rate and extent of ATP decrease. Each preconditioning attenuated the stress-induced insolubility among total cellular protein as well as the inactivation and insolubilization of cytoplasmic and nuclear luciferase. Transient overexpression of human HSP70 in cells also attenuated both the cytotoxic and proteotoxic effects of ATP depletion. Quercetin, a blocker of stress-responsive HSP expression, abolished the effects of stressful preconditioning but did not influence the effects of overexpressed HSP70. Analyses of the cellular fractions revealed that both the stress-preconditioned and HSP70-overexpressing cells retain the soluble pool of HSP70 longer during ATP depletion. Larger amounts of other proteins coimmunoprecipitated with excess HSP70 compared with control cells deprived of ATP. This is the first demonstration of positive correlation between chaperone activity within cells and their viability in the context of ischemia-like stress.


Subject(s)
Adenosine Triphosphate/deficiency , HSP70 Heat-Shock Proteins/physiology , Ischemic Preconditioning, Myocardial , Muscle Proteins/physiology , Animals , Cell Death , Cell Line , Cytoprotection , Hot Temperature , Humans , Ischemic Preconditioning, Myocardial/methods , Myocardium/metabolism , Myocardium/pathology , Oxidative Stress , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...