Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters











Publication year range
1.
Bioorg Med Chem ; 26(13): 3825-3836, 2018 07 30.
Article in English | MEDLINE | ID: mdl-30017114

ABSTRACT

Pancreatic cancer poorly responds to available drugs, and finding novel approaches to target this cancer type is of high significance. Here, based on a common property of pancreatic cancer cells to express somatostatin receptors (SSTR), we designed drug conjugates with novel somatostatin-derived cyclic peptides (SSTp) with broad selectivity towards SSTR types to facilitate drug targeting of the pancreatic cancer cells specifically. Uptake of our newly designed SSTps was facilitated by SSTRs expressed in the pancreatic cancers, including SSTR2, SSTR3, SSTR4 and SSTR5. Three major drugs were conjugated to our best SSTps that served as delivery vehicles, including Camptothecin (CPT), Combretastatin-4A (COMB) and Azatoxin (AZA). All designed drug conjugates demonstrated penetration to pancreatic cancer cell lines, and significant toxicity towards them. Furthermore, the drug conjugates specifically accumulated in tumors in the animal xenograft model, though some accumulation was also seen in kidney. Overall these findings lay the basis for development of novel drug series that could target the fatal pancreatic cancer.


Subject(s)
Antineoplastic Agents/chemical synthesis , Peptides, Cyclic/chemistry , Somatostatin/chemistry , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Camptothecin/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Humans , Indoles/chemistry , Kidney/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Peptides, Cyclic/chemical synthesis , Receptors, Somatostatin/antagonists & inhibitors , Receptors, Somatostatin/genetics , Receptors, Somatostatin/metabolism , Stilbenes/chemistry , Tissue Distribution , Xenograft Model Antitumor Assays
2.
Oncogene ; 34(43): 5460-71, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25659585

ABSTRACT

Elevated levels of the inducible heat-shock protein 70 (Hsp72) have been implicated in mammary tumorigenesis in histological investigations of human breast cancer. We therefore examined the role of Hsp72 in mice, using animals in which the hsp70 gene was inactivated. We used a spontaneous tumor system with mice expressing the polyomavirus middle T (PyMT) oncogene under control of the mouse mammary tumor virus (MMTV) long-terminal repeat (MMT mice). These mice developed spontaneous, metastatic mammary cancer. We then showed Hsp72 to be upregulated in a fraction of mammary cancer initiating cells (CIC) within the MMT tumor cell population. These cells were characterized by elevated surface levels of stem cell markers CD44 and Sca1 and by rapid metastasis. Inactivation of the hsp70 gene delayed the initiation of mammary tumors. This delay in tumor initiation imposed by loss of hsp70 was correlated with a decreased pool of CIC. Interestingly, hsp70 knockout significantly reduced invasion and metastasis by mammary tumor cells and implicated its product Hsp72 in cell migration and formation of secondary neoplasms. Impaired tumorigenesis and metastasis in hsp70-knockout MMT mice was associated with downregulation of the met gene and reduced activition of the oncogenic c-Met protein. These experiments therefore showed Hsp72 to be involved in the growth and progression of mammary carcinoma and highlighted this protein as a potential target for anticancer drug development.


Subject(s)
Cell Transformation, Neoplastic/genetics , HSP72 Heat-Shock Proteins/genetics , Neoplasm Metastasis/genetics , Animals , Antineoplastic Agents/pharmacology , Cell Movement/drug effects , Cell Movement/genetics , Cell Transformation, Neoplastic/drug effects , Down-Regulation/drug effects , Down-Regulation/genetics , Female , Mammary Neoplasms, Experimental/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Metastasis/drug therapy , Oncogenes/genetics , Proto-Oncogene Proteins c-met/genetics
3.
Oncogene ; 34(32): 4153-61, 2015 Aug 06.
Article in English | MEDLINE | ID: mdl-25347739

ABSTRACT

Mechanistic studies from cell culture and animal models have revealed critical roles for the heat shock protein Hsp70 in cancer initiation and progression. Surprisingly, many effects of Hsp70 on cancer have not been related to its chaperone activity, but rather to its role(s) in regulating cell signaling. A major factor that directs Hsp70 signaling activity appears to be the co-chaperone Bag3. Here, we review these recent breakthroughs, and how these discoveries drive drug development efforts.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , HSP70 Heat-Shock Proteins/metabolism , Neoplasms/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis Regulatory Proteins/genetics , Gene Expression Regulation, Neoplastic , HSP70 Heat-Shock Proteins/genetics , Humans , Models, Biological , Neoplasms/genetics , Protein Binding
4.
Oncogene ; 33(2): 212-24, 2014 Jan 09.
Article in English | MEDLINE | ID: mdl-23246965

ABSTRACT

Reduced expression of the p53 family member p63 has been suggested to play a causative role in cancer metastasis. Here, we show that ΔNp63α, the predominant p63 isoform, plays a major role in regulation of cell migration, invasion and cancer metastasis. We identified mitogen-activated protein (MAP) kinase phosphatase 3 (MKP3) as a downstream target of ΔNp63α that is required for mediating these effects. We show that ΔNp63α regulates extracellular signal-regulated protein kinases 1 and 2 (Erk1/2) activity via MKP3 in both cancer and non-transformed cells. We further show that exogenous ΔNp63α inhibits cell invasion and is dependent on MKP3 upregulation for repression. Conversely, endogenous pan-p63 ablation results in increased cell migration and invasion, which can be reverted by reintroducing the ΔNp63α isoform alone, but not by other isoforms. Interestingly, these effects require Erk2, but not Erk1 expression, and can be rescued by enforced MKP3 expression. Moreover, MKP3 expression is reduced in invasive cancers, and reduced p63 expression increases metastatic frequency in vivo. Taken together, these results suggest an important role for ΔNp63α in preventing cancer metastasis by inhibition of Erk2 signaling via MKP3.


Subject(s)
Dual Specificity Phosphatase 6/physiology , Extracellular Signal-Regulated MAP Kinases/physiology , MAP Kinase Signaling System/physiology , Neoplasm Metastasis/prevention & control , Transcription Factors/physiology , Tumor Suppressor Proteins/physiology , Animals , Cell Line, Tumor , Cell Movement , Female , Humans , Mice , Neoplasm Invasiveness
5.
Oncogene ; 30(25): 2836-45, 2011 Jun 23.
Article in English | MEDLINE | ID: mdl-21297664

ABSTRACT

The major heat shock protein Hsp72 is expressed at elevated levels in many human cancers and its expression correlates with tumor progression. Here, we investigated the role of Hsp72 in Her2 oncogene-induced neoplastic transformation and tumorigenesis. Expression of Her2 in untransformed MCF10A mammary epithelial cells caused transformation, as judged by foci formation in culture and tumorigenesis in xenografts. However, expression of Her2 in Hsp72-depleted cells failed to induce transformation. The anti-tumorigenic effects of Hsp72 downregulation were associated with cellular senescence because of accumulation of p21 and depletion of survivin. Accordingly, either knockdown of p21 or expression of survivin reversed this senescence process. Further, we developed an animal model of Hsp72-dependent breast cancer associated with expression of Her2. Knockout (KO) of Hsp72 almost completely suppressed tumorigenesis in the MMTVneu breast cancer mouse model. In young Hsp72 KO mice, expression of Her2 instead of mammary tissue hyperplasia led to suppression of duct development and blocked alveolar budding. These effects were due to massive cell senescence in mammary tissue, which was associated with upregulation of p21 and downregulation of survivin. Therefore, Hsp72 has an essential role in Her2-induced tumorigenesis by regulating oncogene-induced senescence pathways.


Subject(s)
HSP72 Heat-Shock Proteins/physiology , Mammary Neoplasms, Experimental/physiopathology , Receptor, ErbB-2/physiology , Animals , Blotting, Western , Cell Line, Tumor , Immunohistochemistry , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Knockout , Xenograft Model Antitumor Assays
6.
Oncogene ; 29(37): 5204-13, 2010 Sep 16.
Article in English | MEDLINE | ID: mdl-20622894

ABSTRACT

The heat-shock transcription factor HSF1 was recently shown to have a key role in the development of tumors associated with activation of Ras or inactivation of p53. Here, we show that HSF1 is required for the cell transformation and tumorigenesis induced by the human epidermal growth factor receptor-2 (HER2) oncogene responsible for aggressive breast tumors. Upon expression of HER2, untransformed human mammary epithelial MCF-10A cells underwent neoplastic transformation, formed foci in culture and tumors in nude mouse xenografts. However, expression of HER2 in MCF-10A cells with knockdown of HSF1 did not cause either foci formation or tumor growth in xenografts. The antitumorigenic effect of downregulation of HSF1 was associated with HER2-induced accumulation of the cyclin-dependent kinase inhibitor p21 and decrease in the mitotic regulator survivin, which resulted in growth inhibition and cell senescence. In fact, either knockout of p21 or overexpression of survivin alleviated these effects of HSF1 knockdown. The proliferation of certain human HER2-positive breast cancer lines also requires HSF1, as its knockdown led to upregulation of p21 and/or decrease in survivin, precipitating growth arrest. Similar effects were observed with a small-molecular-weight inhibitor of the heat-shock response NZ28. The effects of HSF1 knockdown on the growth arrest and senescence of HER2-expressing cells were associated with downregulation of heat-shock protein (Hsp)72 and Hsp27. Therefore, HSF1 is critical for proliferation of HER2-expressing cells, most likely because it maintains the levels of HSPs, which in turn control regulators of senescence p21 and survivin.


Subject(s)
Cell Transformation, Neoplastic , DNA-Binding Proteins/physiology , Receptor, ErbB-2/physiology , Transcription Factors/physiology , Animals , Cell Division/physiology , DNA-Binding Proteins/genetics , Gene Knockdown Techniques , Heat Shock Transcription Factors , Humans , Mice , Mice, Nude , Transcription Factors/genetics
7.
Oncogene ; 29(13): 1952-62, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20062073

ABSTRACT

Knockout of heat shock protein Hsp72 was shown to promote chromosomal instability and increase radiation sensitivity of mouse fibroblasts. Here, we report that downregulation of Hsp72 in human tumor cells leads to suppression of a specific branch of the DNA damage response (DDR) that facilitates DNA repair following genotoxic insults, that is, reduced accumulation of the phosphorylated form of histone H2AX (gammaH2AX). This inhibition was due to decreased expression of H2AX as well as higher rate of gammaH2AX dephosphorylation. Formation of gammaH2AX and MDC1 radiation-induced foci was impaired in Hsp72-depleted cells, which in turn enhanced DNA damage, resulting in sensitization of cells to gamma-radiation and doxorubicin. These effects of Hsp72 knockdown were dependent on activation of the p53/p21-signaling pathway. Overall, permanent activation of the p53/p21 signaling in Hsp72-depleted cells specifically impaired the gammaH2AX pathway of the DDR, enhanced DNA damage following genotoxic insults, and led to further stimulation of the p53/p21 pathway, thus creating a positive feedback loop. The resulting strong induction of p21 precipitated senescence following exposure to DNA-damaging agents, thus accounting for higher sensitivity of cells to genotoxic stresses.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/physiology , DNA Damage , HSP72 Heat-Shock Proteins/deficiency , Histones/metabolism , Signal Transduction/physiology , Tumor Suppressor Protein p53/physiology , Animals , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/pharmacology , DNA Repair/genetics , Down-Regulation , Gene Knockout Techniques , HSP72 Heat-Shock Proteins/genetics , HSP72 Heat-Shock Proteins/metabolism , Histones/genetics , Humans , Mice , Protein Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins
8.
Vaccine ; 26(26): 3223-6, 2008 Jun 19.
Article in English | MEDLINE | ID: mdl-18467011

ABSTRACT

Aggregation may significantly affect the fate of a polypeptide, including its susceptibility to proteasome-dependent or autophagic degradation, its interaction with chaperones, etc. Since all these factors may affect the antigenicity of a polypeptide, we hypothesized that stimulating aggregation of an antigenic protein by its fusion to polyQ domain may enhance its antigenic potential. This hypothesis was tested with the weakly immunogenic model antigen GFP, which was fused to either long polyQ domain that triggers protein aggregation (103Q), or short polyQ domain that does not promote aggregation (25Q). Plasmids encoding control pGFP or soluble 25Q-GFP generated a very weak antibody response, while a significant increase in anti-GFP antibody titer was seen in groups immunized with DNA encoding aggregating 103Q-GFP. Similarly, fusion with 103Q strongly enhanced anti-GFP CTL activity, compared to fusion with 25Q. No apparent toxicity was observed after immunization with polyQ-GFP fusions. These data suggest that fusion of an antigen with expanded polyQ domains could have a significant adjuvant potential.


Subject(s)
Adjuvants, Immunologic , Immunologic Factors/pharmacology , Peptides/pharmacology , Antibodies/blood , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/immunology , Humans , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology
9.
Int J Hyperthermia ; 21(5): 403-19, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16048838

ABSTRACT

Many major neurodegenerative diseases, including Amyotrophic Lateral Sclerosis, Alzheimer's disease, Parkinson's disease, Huntington Disease and other polyglutamine expansion disorders, are associated with degeneration and death of specific neuronal populations due to accumulation of certain abnormal polypeptides. These misfolded species aggregate and form inclusion bodies and their neurotoxicity is associated with the aggregation. To handle a build-up of abnormal proteins cells employ a complicated machinery of molecular chaperones and various proteolytic systems. Chaperones facilitate refolding or degradation of misfolded polypeptides, prevent protein aggregation and play a role in formation of aggresome, a centrosome-associated body to which small cytoplasmic aggregates are transported. The ubiquitin-proteasome proteolytic system is critical for reducing the levels of soluble abnormal proteins, while autophagy plays the major role in clearing of cells from protein aggregates. Accumulation of the aggregation prone proteins activates signal transduction pathways that control cell death, including JNK pathway that controls viability of a cell in various models of Parkinson's and Huntington's diseases. The major chaperone Hsp72 can interfere with this signalling pathway, thus promoting survival. A very important consequence of a build-up and aggregation of misfolded proteins is impairment of the ubiquitin-proteasome degradation system and suppression of the heat shock response. Such an inhibition of the major cell defense systems may play a critical role in neurodegeneration. Here, it is suggested that these changes may reflect a senescence-like programme initiated by the aggregated abnormal polypeptides. Pathways that control the fate of misfolded proteins, for example molecular chaperones or proteolytic systems, may become interesting novel targets for therapy of neurodegenerative disorders.


Subject(s)
Molecular Chaperones/physiology , Neurodegenerative Diseases/physiopathology , Apoptosis/physiology , Cellular Senescence/physiology , Heat-Shock Proteins/physiology , Humans , Inclusion Bodies/metabolism , Models, Biological , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/therapy , Protein Conformation , Protein Folding , Ubiquitin-Protein Ligases/physiology
10.
Int J Hyperthermia ; 18(3): 203-15, 2002.
Article in English | MEDLINE | ID: mdl-12061330

ABSTRACT

It is now possible to search for new drugs using high-throughput screening of chemical libraries accumulated over the past few years. To detect potential new hyperthermia sensitizers, we are screening for chemical inhibitors of thermotolerance. For the screening of a large chemical library, a rapid and simple assay based on the XTT-tetrazolium salt with the addition of intermediate electron acceptor, phenazine methosulphate (PMS) as a promoter, was developed. It was found that the sensitivity of the XTT/PMS assay is sufficient for assessing thermal cell killing and thermotolerance, although it was highly dependent on cell number and type. When the formazan assay system was challenged with the bioflavonoid drug quercetin (up to 25mm) and validated against the clonogenic cell survival assay, significant decreases in thermotolerant cell viability were observed, directly reflecting inhibition of thermotolerance. Although short-term assays can, in some instances, underestimate overall cell killing, the dose dependency of inhibition of thermotolerance by quercetin recorded in this study by clonogenic and XTT/PMS assays was similar. Application of the XTT/PMS assay in chemical library screening was highly effective in differentiating potential thermotolerance inhibitors from both chemicals with lack of efficacy and from toxic compounds. Taken together, these results show that the XTT/PMS assay, when carried out under careful conditions, is well suited for primary high-flux screen of many thousands of compounds, thus opening up new areas for discovery of hyperthermia sensitizers.


Subject(s)
Hyperthermia, Induced , Tetrazolium Salts , 3T3 Cells , Animals , Cell Survival/drug effects , Colony-Forming Units Assay , Drug Evaluation, Preclinical , Hot Temperature , Methylphenazonium Methosulfate , Mice , Quercetin/pharmacology
11.
J Cell Biol ; 153(4): 851-64, 2001 May 14.
Article in English | MEDLINE | ID: mdl-11352944

ABSTRACT

Abnormal proteins, which escape chaperone-mediated refolding or proteasome-dependent degradation, aggregate and form inclusion bodies (IBs). In several neurodegenerative diseases, such IBs can be formed by proteins with expanded polyglutamine (polyQ) domains (e.g., huntingtin). This work studies the regulation of intracellular IB formation using an NH(2)-terminal fragment of huntingtin with expanded polyQ domain. We demonstrate that the active form of MEKK1, a protein kinase that regulates several stress-activated signaling cascades, stimulates formation of the IBs. This function of MEKK1 requires kinase activity, as the kinase-dead mutant of MEKK1 cannot stimulate this process. Exposure of cells to UV irradiation or cisplatin, both of which activate MEKK1, also augmented the formation of IBs. The polyQ-containing huntingtin fragment exists in cells in two distinct forms: (a) in a discrete soluble complex, and (b) in association with insoluble fraction. MEKK1 strongly stimulated recruitment of polyQ polypeptides into the particulate fraction. Notably, a large portion of the active form of MEKK1 was associated with the insoluble fraction, concentrating in discrete sites, and polyQ-containing IBs always colocalized with them. We suggest that MEKK1 is involved in a process of IB nucleation. MEKK1 also stimulated formation of IBs with two abnormal polypeptides lacking the polyQ domain, indicating that this kinase has a general effect on protein aggregation.


Subject(s)
Inclusion Bodies/enzymology , MAP Kinase Kinase Kinase 1 , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Peptides/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Cytosol/enzymology , HeLa Cells , Heat-Shock Response/physiology , Hippocampus/cytology , Humans , Huntingtin Protein , Inclusion Bodies/chemistry , Kidney/cytology , MAP Kinase Signaling System/physiology , Mutagenesis/physiology , Nerve Tissue Proteins/chemistry , Neurons/cytology , Neurons/radiation effects , Nuclear Proteins/chemistry , Protein Structure, Tertiary , Rats , Solubility , Transfection , Ultraviolet Rays
13.
J Biol Chem ; 276(6): 3920-8, 2001 Feb 09.
Article in English | MEDLINE | ID: mdl-11062236

ABSTRACT

In addition to promoting protein folding and translocation, molecular chaperones of Hsp70/DnaJ families are essential for the selective breakdown of many unfolded proteins. It has been proposed that chaperones function in degradation to maintain the substrates in a soluble form. In Escherichia coli, a nonsecreted alkaline phosphatase mutant that lacks its signal sequence (PhoADelta2-22) fails to fold in the cytosol and is rapidly degraded at 37 degrees C. We show that PhoADelta2-22 is degraded by two ATP-dependent proteases, La (Lon) and ClpAP, and breakdown by both is blocked in a dnaJ259-ts mutant at 37 degrees C. Both proteases could be immunoprecipitated with PhoA, but to a much lesser extent in the dnaJ mutant. Therefore, DnaJ appears to promote formation of protease-substrate complexes. DnaJ could be coimmunoprecipitated with PhoA, and the extent of this association directly correlated with its rate of degradation. Although PhoA was not degraded when DnaJ was inactivated, 50% or more of the PhoA remained soluble. PhoA breakdown and solubility did not require ClpB. PhoA degradation was reduced in a thioredoxin-reductase mutant (trxB), which allowed PhoADelta2-22 to fold into an active form in the cytosol. Introduction of the dnaJ mutation into trxB cells further stabilized PhoA, increased enzyme activity, and left PhoA completely soluble. Thus, DnaJ, although not necessary for folding (or preventing PhoA aggregation), is required for PhoA degradation and must play an active role in this process beyond maintaining the substrate in a soluble form.


Subject(s)
Bacterial Proteins/metabolism , Escherichia coli/metabolism , Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Adenosine Triphosphate/metabolism , Escherichia coli Proteins , HSP40 Heat-Shock Proteins , Hydrolysis , Protein Folding , Solubility
14.
Mol Cell Biol ; 20(19): 7146-59, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10982831

ABSTRACT

Cellular stress can trigger a process of self-destruction known as apoptosis. Cells can also respond to stress by adaptive changes that increase their ability to tolerate normally lethal conditions. Expression of the major heat-inducible protein hsp70 protects cells from heat-induced apoptosis. hsp70 has been reported to act in some situations upstream or downstream of caspase activation, and its protective effects have been said to be either dependent on or independent of its ability to inhibit JNK activation. Purified hsp70 has been shown to block procaspase processing in vitro but is unable to inhibit the activity of active caspase 3. Since some aspects of hsp70 function can occur in the absence of its chaperone activity, we examined whether hsp70 lacking its ATPase domain or the C-terminal EEVD sequence that is essential for peptide binding was required for the prevention of apoptosis. We generated stable cell lines with tetracycline-regulated expression of hsp70, hsc70, and chaperone-defective hsp70 mutants lacking the ATPase domain or the C-terminal EEVD sequence or containing AAAA in place of EEVD. Overexpression of hsp70 or hsc70 protected cells from heat shock-induced cell death by preventing the processing of procaspases 9 and 3. This required the chaperone function of hsp70 since hsp70 mutant proteins did not prevent procaspase processing or provide protection from apoptosis. JNK activation was inhibited by both hsp70 and hsc70 and by each of the hsp70 domain mutant proteins. The chaperoning activity of hsp70 is therefore not required for inhibition of JNK activation, and JNK inhibition was not sufficient for the prevention of apoptosis. Release of cytochrome c from mitochondria was inhibited in cells expressing full-length hsp70 but not in cells expressing the protein with ATPase deleted. Together with the recently identified ability of hsp70 to inhibit cytochrome c-mediated procaspase 9 processing in vitro, these data demonstrate that hsp70 can affect the apoptotic pathway at the levels of both cytochrome c release and initiator caspase activation and that the chaperone function of hsp70 is required for these effects.


Subject(s)
Apoptosis/physiology , HSP70 Heat-Shock Proteins/physiology , Protein Folding , Stress, Physiological/metabolism , Adaptation, Physiological , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/deficiency , Adenosine Triphosphatases/genetics , Amino Acid Sequence , Amino Acid Substitution , Carrier Proteins/physiology , Caspase 3 , Caspase 9 , Caspases/metabolism , Cell Division , Cell Line , Cytochrome c Group/metabolism , Enzyme Activation , Enzyme Precursors/metabolism , HSC70 Heat-Shock Proteins , HSP70 Heat-Shock Proteins/chemistry , HSP70 Heat-Shock Proteins/deficiency , HSP70 Heat-Shock Proteins/genetics , Hot Temperature , Humans , JNK Mitogen-Activated Protein Kinases , Mitochondria/enzymology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/physiology , Signal Transduction , Stress, Physiological/pathology , Structure-Activity Relationship , Transfection
15.
J Biol Chem ; 275(48): 38088-94, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-10978340

ABSTRACT

Since protection of cells from stress-induced apoptosis by the heat shock protein Hsp72 involves suppression of stress kinase JNK, we suggested that Hsp72-mediated JNK inhibition might also be critical for myocardial protection from ischemia/reperfusion. Transient energy deprivation of H9c2 myogenic cells, used as an in vitro model of myocardial ischemia, led to cell death that had morphological features of apoptosis and necrosis and was independent of caspases. Surprisingly, this unusual type of cell death was regulated by JNK and ERK kinases. In fact, specific inhibition of JNK increased cell survival; specific inhibition of ERKs enhanced deleterious consequences of energy deprivation, whereas inhibition of p38 kinase had no effect. Hsp72 suppressed activation of JNK and did not increase ERK activity, suggesting that inhibition of JNK is the important component of Hsp72-mediated protection. Upon transient energy deprivation, activation of JNK proceeds via two distinct pathways, stimulation of JNK phosphorylation by a protein kinase SEK1 and inhibition of JNK dephosphorylation. Remarkably, in cells exposed to transient energy deprivation, Hsp72 enhanced the rate of JNK dephosphorylation but did not affect SEK1 activity. Therefore, it appears that Hsp72 specifically down-regulates JNK by accelerating its dephosphorylation, which reduces the susceptibility of cardiac cells to simulated ischemia/reperfusion.


Subject(s)
Heat-Shock Proteins/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Muscles/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Death , Cell Line , Enzyme Activation , HSP72 Heat-Shock Proteins , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/metabolism , Muscles/cytology , Phosphorylation , Rats
16.
Mol Cell Biol ; 20(18): 6826-36, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10958679

ABSTRACT

Pretreatment with mild heat shock is known to protect cells from severe stress (acquired thermotolerance). Here we addressed the mechanism of this phenomenon by using primary human fibroblasts. Severe heat shock (45 degrees C, 75 min) of the fibroblasts caused cell death displaying morphological characteristics of apoptosis; however, it was caspase independent. This cell death process was accompanied by strong activation of Akt, extracellular signal-regulated kinase 1 (ERK1) and ERK2, p38, and c-Jun N-terminal (JNK) kinases. Suppression of Akt or ERK1 and -2 kinases increased cell thermosensitivity. In contrast, suppression of stress kinase JNK rendered cells thermoresistant. Development of thermotolerance was not associated with Akt or ERK1 and -2 regulation, and inhibition of these kinases did not reduce acquired thermotolerance. On the other hand, acquired tolerance to severe heat shock was associated with downregulation of JNK. Using an antisense-RNA approach, we found that accumulation of the heat shock protein Hsp72 is necessary for JNK downregulation and is critical for thermotolerance. The capability of naive cells to withstand moderate heat treatment also appears to be dependent on the accumulation of Hsp72 induced by this stress. Indeed, exposure to 45 degrees C for 45 min caused only transient JNK activation and was nonlethal, while prevention of Hsp72 accumulation prolonged JNK activation and led to massive cell death. We also found that JNK activation by UV irradiation, interleukin-1, or tumor necrosis factor was suppressed in thermotolerant cells and that Hsp72 accumulation was responsible for this effect. Hsp72-mediated suppression of JNK is therefore critical for acquired thermotolerance and may play a role in tolerance to other stresses.


Subject(s)
Apoptosis , Caspases/metabolism , Heat-Shock Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Apoptosis/drug effects , Caspase 3 , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/drug effects , HSP72 Heat-Shock Proteins , Heat-Shock Proteins/genetics , Hot Temperature , Humans , JNK Mitogen-Activated Protein Kinases
17.
Cell Stress Chaperones ; 5(2): 139-47, 2000 Apr.
Article in English | MEDLINE | ID: mdl-11147965

ABSTRACT

The major heat shock protein Hsp72 prevents heat-induced apoptosis. We have previously demonstrated that transiently expressed Hsp72 exerts its anti-apoptotic effect by suppressing the activity of stress-kinase JNK, an early component of the apoptotic pathway initiated by heat shock. On the other hand, constitutive expression of Hsp72 does not lead to suppression of heat-induced JNK activation, yet still efficiently prevents apoptosis. To address this apparent contradiction, we studied the effects of constitutively expressed Hsp72 on activation of JNK and apoptosis in Rat-1 fibroblasts. We found that the level of heat-induced apoptosis directly correlated with the duration rather than the magnitude of JNK activity following heat shock. Constitutively expressed Hsp72 strongly reduced the duration of JNK while it did not suppress initial JNK activation. These effects were due to Hsp72-mediated acceleration of JNK dephosphorylation. Addition of vanadate to inhibit JNK phosphatase activity completely prevented the anti-apoptotic action of Hsp72. Therefore, suppression of heat-induced apoptosis by Hsp72 could be fully accounted for by its effects on JNK activity.


Subject(s)
Apoptosis , Heat-Shock Proteins/metabolism , Heat-Shock Response , Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Line , Enzyme Activation , Fibroblasts/cytology , HSP72 Heat-Shock Proteins , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Phosphorylation , Rats
18.
FEBS Lett ; 461(1-2): 73-6, 1999 Nov 12.
Article in English | MEDLINE | ID: mdl-10561499

ABSTRACT

A major inducible heat shock protein, Hsp72, has previously been found to stimulate dephosphorylation (inactivation) of stress kinase JNK in heat-shocked cells and protect them from apoptosis. Using Rat-1 fibroblasts with constitutive expression of a human Hsp72 or its deletion mutant lacking an ATPase domain (C-terminal fragment (CTF)), we tested whether ATPase activity of Hsp72 is necessary for these effects. We found that expression of CTF markedly increased, similarly to the intact protein, JNK dephosphorylation in heat-shocked cells. As a result, JNK inactivation following heat shock occurred much faster in cells expressing either full-length or mutant Hsp72 than in parental cells and this was accompanied by suppression of heat-induced apoptosis. Thus, protein refolding activity of Hsp72 appears to be dispensable for its effect on JNK inactivation and apoptosis.


Subject(s)
Adenosine Triphosphatases/metabolism , Apoptosis , Heat-Shock Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Line , Fibroblasts/metabolism , HSP72 Heat-Shock Proteins , Humans , Immunoblotting , JNK Mitogen-Activated Protein Kinases , Phosphorylation , Rats , Recombinant Proteins/metabolism , Signal Transduction , Temperature , Time Factors
19.
J Biol Chem ; 274(29): 20223-8, 1999 Jul 16.
Article in English | MEDLINE | ID: mdl-10400639

ABSTRACT

Activation of the c-Jun N-terminal kinase (JNK) by a variety of stimuli is critical for regulation of many cellular processes including apoptosis. The major inducible heat shock protein Hsp72 has previously been demonstrated to inhibit activation of JNK in cells exposed to heat shock and other protein-damaging agents, thus suppressing apoptosis. Hsp72 can protect proteins from stress-induced damage. To test if this protective function of Hsp72 is involved in JNK suppression, we investigated whether Hsp72 can avert activation of JNK by stimuli that do not cause protein damage. We show that Hsp72 suppresses activation of JNK induced by non-protein-damaging stimuli, interleukin-1 and UV irradiation, as well as by constitutively active components of the JNK signaling cascade Cdc42 and MEKK1. Furthermore, Hsp72 strongly reduced activation of JNK by phosphatase inhibitors. We also demonstrate that an Hsp72 mutant that lacks the ATPase domain is still capable of JNK suppression, thus indicating that the protein refolding activity of Hsp72 is not critical for inhibition of JNK activation. Taken together these data suggest that Hsp72 plays a regulatory role in JNK signaling and that the function of Hsp72 in protein protection or refolding is not involved in JNK regulation.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Heat-Shock Proteins/metabolism , Mitogen-Activated Protein Kinases , Adenosine Triphosphatases/metabolism , Animals , COS Cells , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Enzyme Activation , HSP72 Heat-Shock Proteins , Heat-Shock Proteins/drug effects , Heat-Shock Proteins/genetics , Heat-Shock Proteins/radiation effects , Heat-Shock Response , Interleukin-1/pharmacology , JNK Mitogen-Activated Protein Kinases , Protein Folding , Ultraviolet Rays
20.
Oncogene ; 18(24): 3648-51, 1999 Jun 17.
Article in English | MEDLINE | ID: mdl-10380887

ABSTRACT

Hsp72 is the major heat shock-inducible protein capable of protecting cells from a variety of stresses. In non-transformed cells at normal conditions Hsp72 is expressed at very low levels. It is, however, present at elevated levels in the major fraction of tumors and in many transformed cell lines. It is commonly assumed that in tumor cells the expression of Hsp72 at elevated levels is the consequence of oncogenic transformation. In the present study we addressed an alternative possibility that Hsp72 plays an active role in the process of oncogenic transformation. We report here that when Hsp72 was expressed in the Rat-1 fibroblasts either constitutively or from an adenovirus-based construct, cells become oncogenically transformed by the following criteria: loss of contact inhibition and formation of foci characteristic for oncogenically transformed cells; acquisition of the ability to grow in an anchorage-independent manner and to form colonies in soft agar; generation of tumors upon injection into mice. Furthermore, we also report that turning off the Hsp72 expression led to the reversal of the transformed phenotype. We also show that oncogenic potential of Hsp72 is confined in its peptide binding domain since the expression of this domain alone was sufficient for oncogenic transformation of Rat-1 cells.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Heat-Shock Proteins/metabolism , Oncogene Proteins/metabolism , Adenoviridae/genetics , Animals , Binding Sites , Cell Line , Cell Transformation, Neoplastic/genetics , Contact Inhibition , Gene Expression/drug effects , HSP72 Heat-Shock Proteins , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/genetics , Mice , Mice, Nude , Neoplasm Transplantation , Oncogene Proteins/chemistry , Oncogene Proteins/genetics , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phenotype , Rats , Tetracycline/pharmacology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL