Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Int J Mol Sci ; 22(20)2021 Oct 19.
Article in English | MEDLINE | ID: mdl-34681913

ABSTRACT

Histone deacetylase inhibitors (HDACis) are one of the therapeutic options for cutaneous T-cell lymphoma (CTCL), but they have limited effects. We previously demonstrated that HSP72 overexpression is associated with chemoresistance to HDACis in lymphoma cells. The purpose of this study was to investigate whether the functional depletion of HSP72 enhances the effect of the HDACi vorinostat. First, we established a stable HSP72-knockdown CTCL cell line and confirmed the influence of HSP72 reduction on the antitumor effects of vorinostat. Next, we studied the effect of quercetin, an inhibitor of HSP72, on the antineoplastic effects of vorinostat. In five CTCL cell lines examined, HSP72 expression was highest in Hut78 cells, and HSP72 knockdown enhanced vorinostat-induced apoptosis in these cells. Low-dose quercetin reduced HSP72 expression, increased HDAC activity, and enhanced vorinostat-induced suppression of Hut78 cell proliferation. A single low dose of quercetin induced G2 arrest and only slightly increased the sub-G1 cell fraction. Quercetin also significantly enhanced vorinostat-induced apoptosis, caspase-3, caspase-8, and caspase-9 activity, and the loss of mitochondrial membrane potential. HSP72 knockdown enhanced vorinostat-induced apoptosis in an HSP72-overexpressing CTCL cell line, and thus, quercetin may be a suitable candidate for combination therapy with vorinostat in clinical settings.


Subject(s)
HSP72 Heat-Shock Proteins/genetics , Lymphoma, T-Cell, Cutaneous/genetics , Quercetin/pharmacology , RNA, Small Interfering/pharmacology , Skin Neoplasms/genetics , Vorinostat/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , HSP72 Heat-Shock Proteins/antagonists & inhibitors , Histone Deacetylases/metabolism , Humans , Lymphoma, T-Cell, Cutaneous/drug therapy , Skin Neoplasms/drug therapy
2.
Molecules ; 25(18)2020 Sep 16.
Article in English | MEDLINE | ID: mdl-32947765

ABSTRACT

Covalent inhibition has recently gained a resurgence of interest in several drug discovery areas. The expansion of this approach is based on evidence elucidating the selectivity and potency of covalent inhibitors when bound to particular amino acids of a biological target. The unexpected covalent inhibition of heat shock protein 72 (HSP72) by covalently targeting Lys-56 instead of Cys-17 was an interesting observation. However, the structural basis and conformational changes associated with this preferential coupling to Lys-56 over Cys-17 remain unclear. To resolve this mystery, we employed structural and dynamic analyses to investigate the structural basis and conformational dynamics associated with the unexpected covalent inhibition. Our analyses reveal that the coupling of the irreversible inhibitor to Lys-56 is intrinsically less dynamic than Cys-17. Conformational dynamics analyses further reveal that the coupling of the inhibitor to Lys-56 induced a closed conformation of the nucleotide-binding subdomain (NBD) α-helices, in contrast, an open conformation was observed in the case of Cys-17. The closed conformation maintained the crucial salt-bridge between Glu-268 and Lys-56 residues, which strengthens the interaction affinity of the inhibitor nearly identical to adenosine triphosphate (ADP/Pi) bound to the HSP72-NBD. The outcome of this report provides a substantial shift in the conventional direction for the design of more potent covalent inhibitors.


Subject(s)
Cysteine/chemistry , HSP72 Heat-Shock Proteins/metabolism , Lysine/chemistry , Adenosine/chemistry , Cluster Analysis , HSP72 Heat-Shock Proteins/antagonists & inhibitors , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Principal Component Analysis , Protein Conformation, alpha-Helical
3.
J Med Chem ; 62(24): 11383-11398, 2019 12 26.
Article in English | MEDLINE | ID: mdl-31725295

ABSTRACT

The covalent inhibition mechanism of action, which overcomes competition with high-affinity, high-abundance substrates of challenging protein targets, can deliver effective chemical probes and drugs. The success of this strategy has centered on exposed cysteine residues as nucleophiles but the low abundance of cysteine in the proteome has limited its application. We have recently reported our discovery that lysine-56 in the difficult-to-drug target HSP72 could form a covalent bond with a small-molecule inhibitor. We now disclose the optimization of these targeted covalent inhibitors using rational design. Essential to our optimization was the development of a new covalent fluorescence polarization assay, which allows for the direct measurement of the key kinetic parameter in covalent inhibitor design, kinact/KI, extrapolation of the underlying parameters, kinact and Ki, and direct comparison to reversible analogues. Using our approach, we demonstrate a >100-fold enhancement in covalent efficiency and key learnings in lysine-selective electrophile optimization.


Subject(s)
Drug Discovery , HSP72 Heat-Shock Proteins/antagonists & inhibitors , Lysine/chemistry , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Humans , Kinetics , Molecular Structure , Structure-Activity Relationship
4.
Oncogene ; 38(4): 469-482, 2019 01.
Article in English | MEDLINE | ID: mdl-30166590

ABSTRACT

TNFα is a pleiotropic cytokine which fuels tumor cell growth, invasion, and metastasis in some malignancies, while in others it induces cytotoxic cell death. However, the molecular mechanism by which TNFα exerts its diverse effects on breast cancer subtypes remains elusive. Using in vitro assays and mouse xenografts, we show here that TNFα contributes to the aggressive properties of triple negative breast cancer (TNBC) cell lines via upregulation of TNFAIP3(A20). In a striking contrast, TNFα induces a potent cytotoxic cell death in luminal (ER+) breast cancer cell lines which fail to upregulate A20 expression. Overexpression of A20 not only protects luminal breast cancer cell lines from TNFα-induced cell death via inducing HSP70-mediated anti-apoptotic pathway but also promotes a robust EMT/CSC phenotype by activating the pStat3-mediated inflammatory signaling. Furthermore, A20 overexpression in luminal breast cancer cells induces aggressive metastatic properties in mouse xenografts via generating a permissive inflammatory microenvironment constituted by granulocytic-MDSCs. Collectively, our results reveal a mechanism by which A20 mediates pleiotropic effects of TNFα playing role in aggressive behaviors of TNBC subtype while its deficiency results in TNFα-induced apoptotic cell death in luminal breast cancer subtype.


Subject(s)
Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Genetic Pleiotropy , Neoplasm Proteins/physiology , Tumor Necrosis Factor alpha-Induced Protein 3/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Apoptosis/physiology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Female , HSP72 Heat-Shock Proteins/antagonists & inhibitors , HSP72 Heat-Shock Proteins/physiology , Heterografts , Humans , Inflammation , Lung Neoplasms/secondary , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness/genetics , Neoplasm Proteins/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Purine Nucleosides/pharmacology , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , STAT3 Transcription Factor/physiology , Signal Transduction , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Tumor Necrosis Factor alpha-Induced Protein 3/biosynthesis , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Tumor Necrosis Factor-alpha/genetics
5.
Oncol Rep ; 40(2): 895-901, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29901164

ABSTRACT

Curcumin has been revealed to inhibit liver cancer, however, no studies have reported that the mechanism of curcumin's action on liver cancer is related to damage-associated molecular pattern (DAMP) molecules heat shock protein 70 (HSP70) and the toll-like receptor 4 (TLR4) signaling. This study aimed to investigate whether the activation of TLR4 signaling by HSP70 could be inhibited by curcumin, thus investigating the possible mechanism of curcumin in the inhibition of liver cancer. Western blotting was used to evaluate the expression of the HSP70 and TLR4 in HepG2 cells and ELISA was used to detect the concentration of HSP70 in cell culture medium. A thermal tolerance HepG2 (HepG2TT) cell model was established to simulate HSP70 accumulation in the microenvironment. A certain concentration of curcumin was co-cultured with HepG2 and HepG2TT cells to observe the changes of HSP70 and TLR4. Our results revealed that heat stress significantly increased the expression of extracellular HSP70 (eHSP70) and TLR4 (P<0.01), but significantly reduced the expression of intracellular HSP70 (P<0.01). Curcumin inhibited proliferation, invasion, and metastasis of HepG2 cells, caused cells to remain in the DNA S phase, promoted apoptosis, and significantly reduced intracellular HSP70, eHSP70 and TLR4 levels of HepG2TT cells. Following the removal of curcumin, eHSP70 increased again. In summary, our results demonstrated that the antitumor effect of curcumin was related to the inhibition HSP70-TLR4 signaling.


Subject(s)
Alarmins/antagonists & inhibitors , Curcumin/pharmacology , HSP72 Heat-Shock Proteins/antagonists & inhibitors , Liver Neoplasms/drug therapy , Neoplasm Metastasis/drug therapy , Toll-Like Receptor 4/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Neoplasm Invasiveness/genetics , S Phase/drug effects , Tumor Microenvironment/drug effects
6.
Antiviral Res ; 146: 21-27, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28807685

ABSTRACT

Ebola virus (EBOV) causes a severe disease in humans with the potential for significant international public health consequences. Currently, treatments are limited to experimental vaccines and therapeutics. Therefore, research into prophylaxis and antiviral strategies to combat EBOV infections is of utmost importance. The requirement for high containment laboratories to study EBOV infection is a limiting factor for conducting EBOV research. To overcome this issue, minigenome systems have been used as valuable tools to study EBOV replication and transcription mechanisms and to screen for antiviral compounds at biosafety level 2. The most commonly used EBOV minigenome system relies on the ectopic expression of the T7 RNA polymerase (T7), which can be limiting for certain cell types. We have established an improved EBOV minigenome system that utilizes endogenous RNA polymerase II (pol II) as a driver for the synthesis of minigenome RNA. We show here that this system is as efficient as the T7-based minigenome system, but works in a wider range of cell types, including biologically relevant cell types such as bat cells. Importantly, we were also able to adapt this system to a reliable and cost-effective 96-well format antiviral screening assay with a Z-factor of 0.74, indicative of a robust assay. Using this format, we identified JG40, an inhibitor of Hsp70, as an inhibitor of EBOV replication, highlighting the potential for this system as a tool for antiviral drug screening. In summary, this updated EBOV minigenome system provides a convenient and effective means of advancing the field of EBOV research.


Subject(s)
Antiviral Agents/pharmacology , Ebolavirus/drug effects , Ebolavirus/genetics , Genome, Viral , Microbial Sensitivity Tests/methods , RNA Polymerase II/genetics , Animals , Antiviral Agents/isolation & purification , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Ebolavirus/enzymology , HSP72 Heat-Shock Proteins/antagonists & inhibitors , Hemorrhagic Fever, Ebola/virology , High-Throughput Screening Assays/economics , High-Throughput Screening Assays/instrumentation , High-Throughput Screening Assays/methods , Humans , Microbial Sensitivity Tests/economics , Microbial Sensitivity Tests/instrumentation , RNA Polymerase II/metabolism , RNA, Viral/genetics , Transcription, Genetic/drug effects , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication/drug effects
7.
Amino Acids ; 48(2): 499-504, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26433893

ABSTRACT

The aim of this study was to investigate if a pre-exercise alkalosis-mediated attenuation of HSP72 had any effect on the response of the same stress protein after a subsequent exercise. Seven physically active males [25.0 ± 6.5 years, 182.1 ± 6.0 cm, 74.0 ± 8.3 kg, peak aerobic power (PPO) 316 ± 46 W] performed a repeated sprint exercise (EXB1) following a dose of 0.3 g kg(-1) body mass of sodium bicarbonate (BICARB), or a placebo of 0.045 g kg(-1) body mass of sodium chloride (PLAC). Participants then completed a 90-min intermittent cycling protocol (EXB2). Monocyte expressed HSP72 was significantly attenuated after EXB1 in BICARB compared to PLAC, however, there was no difference in the HSP72 response to the subsequent EXB2 between conditions. Furthermore there was no difference between conditions for measures of oxidative stress (protein carbonyl and HSP32). These findings confirm the sensitivity of the HSP72 response to exercise-induced changes in acid-base status in vivo, but suggest that the attenuated response has little effect upon subsequent stress in the same day.


Subject(s)
Alkalosis/pathology , Exercise/physiology , HSP72 Heat-Shock Proteins/metabolism , Sodium Bicarbonate/pharmacology , Adult , HSP72 Heat-Shock Proteins/antagonists & inhibitors , Humans , Male , Oxidative Stress , Sodium Chloride/metabolism
8.
Nutrition ; 30(10): 1185-94, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24976418

ABSTRACT

OBJECTIVE: L-Alanyl-glutamine (L-Ala-Gln) is a pharmaco-nutrient commonly used in nutrition regimens due to its immunomodulatory effects. In critically ill patients who are septic, L-Ala-Gln was associated with an increase in mortality. The aim of this study was to investigate whether L-Ala-Gln modulated heat shock protein (Hsp)-72, 90-α, T helper (Th)1, Th2, and Th17 cytokine expression in the peripheral blood mononuclear cells (PBMC) of patients with severe sepsis. METHODS: Time-dose effects of L-Ala-Gln were compared with those of L-glutamine (L-Gln) and lipopolysaccharide (LPS) and to healthy controls. PBMCs were incubated with 1 or 10 µg/mL LPS, 5 or 10 mM L-Gln, and 5 or 10 mM L-Ala-Gln for different periods of time (0; 4; 24 h) when culture supernatants were harvested. RESULTS: In both groups, basal Hsp72 increased over time (P < 0.02); Hsp90-α levels declined in controls (P < 0.02) but remained increased in septic patients (P < 0.02), not exhibiting any significant time-response trend. Both Glns suppressed Hsp72 in septic and controls at 10 mM by 4 h (P < 0.045) and Hsp90-α in the control group by 24 h (P < 0.045). LPS did not induce Hsps in either group. L-Ala-Gln did not induce any of the Th1, Th2, and Th17 cytokines in either group. CONCLUSION: High doses of L-Gln or L-Ala-Gln do not induce any of the Th1, Th2, and Th17 cytokines in either healthy or septic human PBMCs. High Gln doses suppress Hsp72 in septic and control PBMCs. Hsp90-α time-series expression declines, contrasting the increasing trend of Hsp72 in healthy controls. Hsp90-α sustains increased levels in septic supernatants, showing a characteristic longitudinal behavior needed further elucidation.


Subject(s)
Cytokines/metabolism , Glutamine/pharmacology , Heat-Shock Proteins/antagonists & inhibitors , Leukocytes, Mononuclear/drug effects , Sepsis/immunology , Dipeptides/pharmacology , HSP72 Heat-Shock Proteins/antagonists & inhibitors , Humans , Leukocytes, Mononuclear/metabolism , Lipopolysaccharides , Th1 Cells/metabolism , Th17 Cells/metabolism , Th2 Cells/metabolism
9.
Acta Biochim Pol ; 61(2): 379-83, 2014.
Article in English | MEDLINE | ID: mdl-24904931

ABSTRACT

Ischemic stroke is the second leading cause of death worldwide. One of the main risk factors of the ischemic stroke is atherosclerosis which is a chronic inflammatory and immune-mediated disease. Bacterial infections generate specific human antibodies against various antigens, including Hsps. It has been demonstrated that Hsps are selectively overexpressed in the atherosclerotic lesions. The amino acid sequence homology between human and bacterial Hsps may lead to an autoimmune response by immunological cross-reaction. Such immune response against Hsps overexpressed in the blood vessels under stressful conditions may contribute to inflammatory processes and subsequent development of atherosclerosis. In this study we determined the antibody levels against bacterial and human Hsp by ELISA in blood plasma obtained from stroke patients. Using ANOVA we analyzed levels of Hsp-antibodies in control and patient groups and correlate them with several stroke risk factors. The group of stroke patients had elevated levels of anti-Hsp antibodies compared to the control group. We also discovered an antibody level increase in patients that previously underwent another stroke. Our data provide evidence that autoimmunity could underlie formation of atherosclerosis plaque leading to stroke.


Subject(s)
Antibodies, Bacterial/blood , Atherosclerosis/immunology , Autoantibodies/blood , Brain Ischemia/immunology , Stroke/immunology , Adult , Aged , Aged, 80 and over , Antibodies, Bacterial/biosynthesis , Atherosclerosis/blood , Atherosclerosis/etiology , Atherosclerosis/pathology , Autoantibodies/biosynthesis , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/blood , Bacterial Proteins/immunology , Brain Ischemia/blood , Brain Ischemia/complications , Brain Ischemia/pathology , Case-Control Studies , Chaperonin 60/antagonists & inhibitors , Chaperonin 60/blood , Chaperonin 60/immunology , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/blood , Escherichia coli Proteins/immunology , Female , Gene Expression/immunology , HSP40 Heat-Shock Proteins/antagonists & inhibitors , HSP40 Heat-Shock Proteins/blood , HSP40 Heat-Shock Proteins/immunology , HSP70 Heat-Shock Proteins/antagonists & inhibitors , HSP70 Heat-Shock Proteins/blood , HSP70 Heat-Shock Proteins/immunology , HSP72 Heat-Shock Proteins/antagonists & inhibitors , HSP72 Heat-Shock Proteins/blood , HSP72 Heat-Shock Proteins/immunology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/blood , HSP90 Heat-Shock Proteins/immunology , Humans , Male , Middle Aged , Risk Factors , Stroke/blood , Stroke/complications , Stroke/pathology
10.
Int J Oncol ; 45(1): 157-64, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24736948

ABSTRACT

The specific and efficient delivery of small interfering RNA (siRNA) into cancer cells in vivo remains a major obstacle. In this study, we investigated whether ultrasound-targeted microbubble destruction (UTMD) combined with dual targeting of HSP72 and HSC70 in prostate cancer cell lines improve the specific and efficient cell uptake of siRNA, inhibit HSP90 function and induce extensive tumor-specific apoptosis. VCaP cells were transfected with siRNA oligonucleotides. Cell viability assays were used to evaluate the safety of UTMD. The expression of HSP70, HSP90, caspase-8, caspase-3, PARP-1 and cleaved caspase-3 were determined by quantitative PCR and western blotting. Apoptosis and transfection efficiency were detected by flow cytometry. We found that HSP72, HSC70 and HSP90 expression was absent or weak in normal prostate epithelial cells (RWPE-1), and became uniformly and strongly expressed in prostate cancer cells (VCaP). VCaP and RWPE-1 cells expressed very low levels of caspase-8, caspase-3, PARP-1 and cleaved caspase-3. UTMD combined with dual targeting of HSP72 and HSC70 siRNA impoved the efficiency of transfection, cell uptake of siRNA, downregulated HSP70 and HSP90 expression in VCaP cells on the mRNA and protein levels, and upregulated major apoptotic markers (PARP-1, caspase-8, caspase-3 and cleaved caspase-3), thus, inducing extensive tumor-specific apoptosis. The Cell Counting Kit-8 assay showed decreased cellular viability in the HSP72/HSC70-siRNA silenced group. These results suggest that the combination of UTMD with dual targeting of HSP72 and HSC70 may improve the specific and efficient cell uptake of siRNA, inhibit HSP90 function and induce extensive tumor-specific apoptosis, indicating a novel, potential means for targeting therapeutic strategy to prostate cancer cells.


Subject(s)
HSC70 Heat-Shock Proteins/metabolism , HSP72 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/pathology , Apoptosis/drug effects , Cell Line, Tumor , Combined Modality Therapy , Gene Expression Regulation, Neoplastic , HSC70 Heat-Shock Proteins/antagonists & inhibitors , HSP72 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Male , Microbubbles , RNA Interference , Ultrasonography
11.
Am J Physiol Endocrinol Metab ; 306(3): E344-54, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24347059

ABSTRACT

5'-AMP-activated protein kinase (AMPK) plays an important role as a negative regulator of skeletal muscle mass. However, the precise mechanism of AMPK-mediated regulation of muscle mass is not fully clarified. Heat shock proteins (HSPs), stress-induced molecular chaperones, are related with skeletal muscle adaptation, but the association between AMPK and HSPs in skeletal muscle hypertrophy is unknown. Thus, we investigated whether AMPK regulates hypertrophy by mediating HSPs in C2C12 cells. The treatment with AICAR, a potent stimulator of AMPK, decreased 72-kDa HSP (HSP72) expression, whereas there were no changes in the expressions of 25-kDa HSP, 70-kDa heat shock cognate, and heat shock transcription factor 1 in myotubes. Protein content and diameter were less in the AICAR-treated myotubes in those without treatment. AICAR-induced suppression of myotube hypertrophy and HSP72 expression was attenuated in the siRNA-mediated AMPKα knockdown myotubes. AICAR increased microRNA (miR)-1, a modulator of HSP72, and the increase of miR-1 was not induced in AMPKα knockdown condition. Furthermore, siRNA-mediated HSP72 knockdown blocked AICAR-induced inhibition of myotube hypertrophy. AICAR upregulated the gene expression of muscle Ring-finger 1, and this alteration was suppressed in either AMPKα or HSP72 knockdown myotubes. The phosphorylation of p70 S6 kinase Thr(389) was downregulated by AICAR, whereas this was attenuated in AMPKα, but not in HSP72, knockdown myotubes. These results suggest that AMPK inhibits hypertrophy through, in part, an HSP72-associated mechanism via miR-1 and protein degradation pathways in skeletal muscle cells.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , HSP72 Heat-Shock Proteins/physiology , Muscle Fibers, Skeletal/pathology , Ribonucleotides/pharmacology , AMP-Activated Protein Kinases/genetics , Aminoimidazole Carboxamide/pharmacology , Animals , Cells, Cultured , Enzyme Activation/drug effects , Enzyme Activation/physiology , HSP72 Heat-Shock Proteins/antagonists & inhibitors , Hypertrophy/chemically induced , Mice , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Proteolysis/drug effects , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics
12.
Neurotox Res ; 26(1): 64-77, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24366851

ABSTRACT

The aim of the present study was to investigate the effect of sorafenib and quercetin on the induction of apoptosis and autophagy in human anaplastic astrocytoma (MOGGCCM) and glioblastoma multiforme (T98G) cell lines. In MOGGCCM cells, sorafenib initiated mainly apoptosis, mediated by the mitochondrial pathway with mitochondrial membrane permeabilization, cytochrome c release to the cytoplasm, and activation of caspase 9 and 3. Additional incubation with quercetin potentiated the pro-apoptotic properties of sorafenib. In T98G cells, autophagy was observed most frequently after the sorafenib treatment. It was accompanied by increased beclin 1 and LC3II expression. Administration of quercetin after the sorafenib treatment resulted in an increased number of autophagic cells. After simultaneous drug application, the level of autophagy was lower in favour of apoptosis. Inhibition of heat shock proteins expression by specific small interfering RNA significantly increased the sensitivity of both the cell lines to induction of apoptosis, but not autophagy. We demonstrated for the first time that sorafenib and quercetin are very effective programmed cell death inducers in T98G and MOGGCCM cells, especially in cells with blocked expression of heat shock proteins.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Glioma/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Quercetin/pharmacology , Apoptosis/physiology , Astrocytoma/drug therapy , Astrocytoma/physiopathology , Autophagy/physiology , Cell Line, Tumor , Drug Therapy, Combination , Glioblastoma/drug therapy , Glioblastoma/physiopathology , Glioma/physiopathology , HSP27 Heat-Shock Proteins/antagonists & inhibitors , HSP27 Heat-Shock Proteins/metabolism , HSP72 Heat-Shock Proteins/antagonists & inhibitors , HSP72 Heat-Shock Proteins/metabolism , Heat-Shock Proteins , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/physiology , Molecular Chaperones , Necrosis/chemically induced , Necrosis/physiopathology , Niacinamide/pharmacology , Sorafenib
13.
Cell Stress Chaperones ; 19(1): 115-34, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23761196

ABSTRACT

A series of signaling cascades are activated after angiotensin II binds to angiotensin II type I receptor (AT1R), a peptide that is an important mediator of oxidative stress. Hsp70 regulates a diverse set of signaling pathways through interactions with proteins. Here, we tested the hypothesis of angiotensin II AT1R inhibition effect on Hsp70 interaction with Nox4/p22phox complex and Hsp70 leading to actin cytoskeleton modulation in spontaneously hypertensive rats (SHR) vascular smooth muscle cells (VSMCs). SHR and Wistar-Kyotto rats (VSMCs from 8 to 10 weeks) were stimulated with angiotensin II (100 nmol/L) for 15 min (AII), treated with losartan (100 nmol/L) for 90 min (L), and with losartan for 90 min plus angiotensin in the last 15 min (L + AII). Whereas SHR VSMCs exposure to angiotensin II overexpressed AT1R and Nox4 nicotinamide-adenine dinucleotide phosphate (NADPH) oxidase and slightly downregulated caveolin-1 expression, losartan decreased AT1R protein levels and increased caveolin-1 and Hsp70 expression in SHR VSMC membranes. Immunoprecipitation and immunofluorescence confocal microscopy proved interaction and colocalization of membrane translocated Hsp70 and Nox4/p22phox. Increased levels of Hsp70 contrast with the decreased immunoprecipitation of Nox4/p22phox and RhoA in membranes from SHR VSMCs (L) vs SHR VSMCs (AII). Hsp72 depletion resulted in higher Nox4 expression and increased NADPH oxidase activity in VSMCs (L + AII) from SHR when contrasted with nontransfected VSMCs (L + AII). After Hsp72 knockdown in SHR VSMCs, losartan could not impair angiotensin II-enhanced stress fiber formation and focal adhesion assembly. In conclusion, our data showing a negative regulation of Hsp70 on Nox4/p22phox demonstrates a possible mechanism in explaining the antioxidative function joined to cytoskeletal integrity modulation within the effects of losartan in VSMCs from SHR.


Subject(s)
Antihypertensive Agents/pharmacology , HSP70 Heat-Shock Proteins/metabolism , Losartan/pharmacology , Muscle, Smooth, Vascular/drug effects , NADPH Oxidases/metabolism , Angiotensin II/pharmacology , Animals , Caveolin 1/metabolism , Cells, Cultured , Cytoskeleton/drug effects , Cytoskeleton/metabolism , HSP72 Heat-Shock Proteins/antagonists & inhibitors , HSP72 Heat-Shock Proteins/genetics , HSP72 Heat-Shock Proteins/metabolism , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , NADPH Oxidase 4 , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Rats, Wistar , Receptor, Angiotensin, Type 1/chemistry , Receptor, Angiotensin, Type 1/metabolism , rhoA GTP-Binding Protein/metabolism
14.
Toxicol Appl Pharmacol ; 273(3): 580-9, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24126416

ABSTRACT

The aim of the present study was to investigate whether silencing of Hsp27 or Hsp72 expression in glioblastoma multiforme T98G and anaplastic astrocytoma MOGGCCM cells increases their sensitivity to programmed cell death induction upon temozolomide and/or quercetin treatment. Transfection with specific siRNA was performed for the Hsp gene silencing. As revealed by microscopic observation and flow cytometry, the inhibition of Hsp expression was correlated with severe apoptosis induction upon the drug treatment studied. No signs of autophagy were detected. This was correlated with a decreased mitochondrial membrane potential, increased level of cytochrome c in the cytoplasm, and activation of caspase 3 and caspase 9. All these results suggest that the apoptotic signal was mediated by an internal pathway. Additionally, in a large percentage of cells treated with temozolomide, with or without quercetin, granules within the ER system were found, which was accompanied by an increased level of caspase 12 expression. This might be correlated with ER stress. Quercetin and temozolomide also changed the shape of nuclei from circular to "croissant like" in both transfected cell lines. Our results indicate that blocking of Hsp27 and Hsp72 expression makes T98G cells and MOGGCCM cells extremely vulnerable to apoptosis induction upon temozolomide and quercetin treatment and that programmed cell death is initiated by an internal signal.


Subject(s)
Apoptosis/drug effects , Dacarbazine/analogs & derivatives , Gene Silencing , HSP27 Heat-Shock Proteins/genetics , HSP72 Heat-Shock Proteins/genetics , Quercetin/pharmacology , Astrocytoma/drug therapy , Autophagy/drug effects , Caspase 12/genetics , Caspase 12/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Cell Line, Tumor , Cytochromes c/metabolism , Dacarbazine/pharmacology , Endoplasmic Reticulum Stress/drug effects , Glioblastoma/drug therapy , HSP27 Heat-Shock Proteins/antagonists & inhibitors , HSP27 Heat-Shock Proteins/metabolism , HSP72 Heat-Shock Proteins/antagonists & inhibitors , HSP72 Heat-Shock Proteins/metabolism , Heat-Shock Proteins , Humans , Membrane Potential, Mitochondrial/drug effects , Molecular Chaperones , Temozolomide
15.
PLoS One ; 8(7): e69509, 2013.
Article in English | MEDLINE | ID: mdl-23874968

ABSTRACT

The proteasome inhibitor bortezomib (Velcade) is a promising new agent for bladder cancer therapy, but inducible cytoprotective mechanisms may limit its potential efficacy. We used whole genome mRNA expression profiling to study the effects of bortezomib on stress-induced gene expression in a panel of human bladder cancer cell lines. Bortezomib induced strong upregulation of the inducible HSP70 isoforms HSPA1A and HSPA1B isoforms of Hsp72 in 253J B-V and SW780 (HSPA1A(high)) cells, but only induced the HSPA1B isoform in UM-UC10 and UM-UC13 (HSPA1A(low)) cells. Bortezomib stimulated the binding of heat shock factor-1 (HSF1) to the HSPA1A promoter in 253JB-V but not in UM-UC13 cells. Methylation-specific PCR revealed that the HSPA1A promoter was methylated in the HSPA1A(low) cell lines (UM-UC10 and UM-UC13), and exposure to the chromatin demethylating agent 5-aza-2'-deoxycytidine restored HSPA1A expression. Overexpression of Hsp72 promoted bortezomib resistance in the UM-UC10 and UM-UC13 cells, whereas transient knockdown of HSPA1B further sensitized these cells to bortezomib, and exposure to the chemical HSF1 inhibitor KNK-437 promoted bortezomib sensitivity in the 253J B-V cells. Finally, shRNA-mediated stable knockdown of Hsp72 in 253J B-V promoted sensitivity to bortezomib in vitro and in tumor xenografts in vivo. Together, our results provide proof-of-concept for using Hsp72 inhibitors to promote bortezomib sensitivity in bladder cancers and suggest that selective targeting of HSPA1B could produce synthetic lethality in tumors that display HSPA1A promoter methylation.


Subject(s)
Boronic Acids/pharmacology , Cell Death/drug effects , Gene Expression Regulation, Neoplastic/physiology , HSP72 Heat-Shock Proteins/metabolism , Pyrazines/pharmacology , Urinary Bladder Neoplasms/drug therapy , Bortezomib , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA Methylation/genetics , DNA Primers/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , HSP72 Heat-Shock Proteins/antagonists & inhibitors , HSP72 Heat-Shock Proteins/genetics , Humans , Immunoblotting , Microarray Analysis , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
16.
Tumour Biol ; 34(4): 2367-78, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23580181

ABSTRACT

Glioblastoma multiforme is the most aggressive primary brain tumour. At the cellular and molecular levels, several mechanisms responsible for apoptosis or autophagy induction are blocked. Identification of molecular targets stimulating cells to initiate programmed cell death should be performed for therapeutic purposes. A promising solution is the combination of temozolomide and quercetin. The aim of our study was to evaluate the effect of both drugs, applied alone and in combinations, on apoptosis and autophagy induction in human glioblastoma multiforme T98G cells. Our results clearly indicate that quercetin and temozolomide induce apoptosis very significantly, having no effect on autophagy induction. At the molecular level, it was correlated with caspase 3 and 9 activation, cytochrome c release from the mitochondrium and a decrease in the mitochondrial membrane potential. Both drugs are also potent Hsp27 and Hsp72 inhibitors. This suggests that the apoptotic signal goes through an internal pathway. Increased expression of caspase 12 and the presence of several granules in the cytoplasm after temozolomide treatment with or without quercetin preceding appearance of apoptosis may suggest that apoptosis is initiated by ER stress. Additionally, it was accompanied by changes in the nuclear morphology from circular to 'croissant like'.


Subject(s)
Apoptosis/drug effects , Brain Neoplasms/drug therapy , Dacarbazine/analogs & derivatives , Glioblastoma/drug therapy , Quercetin/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Antioxidants/pharmacology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Caspase 12/metabolism , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cytochromes c/metabolism , Dacarbazine/pharmacology , Endoplasmic Reticulum Stress/drug effects , Enzyme Activation , Glioblastoma/metabolism , Glioblastoma/pathology , HSP27 Heat-Shock Proteins/antagonists & inhibitors , HSP72 Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Chaperones , Temozolomide
17.
Mol Cancer Res ; 11(3): 219-29, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23303345

ABSTRACT

The stress-induced HSP70 is an ATP-dependent molecular chaperone that plays a key role in refolding misfolded proteins and promoting cell survival following stress. HSP70 is marginally expressed in nontransformed cells, but is greatly overexpressed in tumor cells. Silencing HSP70 is uniformly cytotoxic to tumor but not normal cells; therefore, there has been great interest in the development of HSP70 inhibitors for cancer therapy. Here, we report that the HSP70 inhibitor 2-phenylethynesulfonamide (PES) binds to the substrate-binding domain of HSP70 and requires the C-terminal helical "lid" of this protein (amino acids 573-616) to bind. Using molecular modeling and in silico docking, we have identified a candidate binding site for PES in this region of HSP70, and we identify point mutants that fail to interact with PES. A preliminary structure-activity relationship analysis has revealed a derivative of PES, 2-(3-chlorophenyl) ethynesulfonamide (PES-Cl), which shows increased cytotoxicity and ability to inhibit autophagy, along with significantly improved ability to extend the life of mice with pre-B-cell lymphoma, compared with the parent compound (P = 0.015). Interestingly, we also show that these HSP70 inhibitors impair the activity of the anaphase promoting complex/cyclosome (APC/C) in cell-free extracts, and induce G2-M arrest and genomic instability in cancer cells. PES-Cl is thus a promising new anticancer compound with several notable mechanisms of action.


Subject(s)
Antineoplastic Agents/administration & dosage , HSP72 Heat-Shock Proteins/antagonists & inhibitors , Neoplasms, Experimental/drug therapy , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Sulfonamides/administration & dosage , Animals , Computer Simulation , Gene Expression Regulation, Leukemic , Genomic Instability/drug effects , HSP72 Heat-Shock Proteins/genetics , HSP72 Heat-Shock Proteins/metabolism , Humans , Mice , Models, Molecular , Molecular Docking Simulation , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Protein Binding/drug effects , Protein Structure, Tertiary/drug effects , Structure-Activity Relationship , Substrate Specificity
18.
Cardiovasc Res ; 96(3): 524-32, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22933322

ABSTRACT

AIMS: Vascular calcification (VC) is a significant contributor to cardiovascular mortality in patients with chronic kidney disease (CKD) and coronary artery disease (CAD). Osteo/chondrocytic transformation and simultaneous dedifferentiation of smooth muscle cells (SMCs) are important in the pathogenesis of VC. Heat-shock protein 72 (HSP72) is a cardioprotective inducible heat-shock protein that functions as a molecular chaperone. However, its role in the development of accelerated vascular dysfunction and calcification is largely unexplored. METHODS AND RESULTS: We describe for the first time marked reduction in HSP72 expression in arteries from patients with CKD and CAD, compared with healthy controls, in vivo. Induction of HSP72 by heat-shock treatment (HST) significantly prevented the development of calcification of human aortic smooth muscle cells (HA-SMCs), in vitro. These anti-calcific effects were abolished following treatment with both quercetin, an HST inhibitor, and HSP72 siRNA knockdown. Induction of HSP72 suppressed Cbfa-1-dependent osteo/chondrocytic transformation and stabilized SMC contractile phenotype through the myocardin-serum response factor (SRF) pathway. Co-immunoprecipitation studies demonstrated physical association between SRF and HSP72. Furthermore, organ culture of arteries from CKD and CAD patients showed that these arteries retained their ability to induce HSP72 following HST, despite initially reduced expression. CONCLUSION: Our study shows for the first time that intracellular HSP72 may function as a central regulator of molecular pathways involved in the development of VC. We suggest treatment strategies that up-regulate HSP72 as a new approach to inhibit VC.


Subject(s)
Coronary Artery Disease/metabolism , HSP72 Heat-Shock Proteins/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Renal Insufficiency, Chronic/metabolism , Vascular Calcification/prevention & control , Adult , Aged , Aged, 80 and over , Case-Control Studies , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/metabolism , Coronary Artery Disease/pathology , Female , HSP72 Heat-Shock Proteins/antagonists & inhibitors , HSP72 Heat-Shock Proteins/genetics , Heat-Shock Response , Humans , Male , Middle Aged , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Nuclear Proteins/metabolism , Organ Culture Techniques , Phenotype , Quercetin/pharmacology , RNA Interference , RNA, Messenger/metabolism , Renal Insufficiency, Chronic/pathology , Serum Response Factor/metabolism , Signal Transduction , Trans-Activators/metabolism , Transfection , Up-Regulation , Vascular Calcification/genetics , Vascular Calcification/metabolism , Vascular Calcification/pathology , Young Adult
19.
Mol Cell Biochem ; 369(1-2): 227-33, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22810500

ABSTRACT

The acute, as well as late, phase of cardioprotection induced by ischemic preconditioning is abolished in hyperlipidemic (HL) rat heart. The pharmacological inhibition of glycogen synthase kinase-3ß (GSK-3ß), has earlier been reported to restore this attenuated acute cardioprotective effect. However, it not known whether GSK-3ß inhibitors administered 24 h before the ischemic injury would restore the late cardioprotective in HL rat and, if yes, the role of heat shock protein 72 (HSP 72) in its modulation. Hyperlipidemia was produced in rat by feeding high-fat diet for 6 weeks. Isolated perfused rat heart was subjected to 30 min of ischemia followed by 120 min of reperfusion (I/R). Myocardial infarct size was estimated by triphenyltetrazolium chloride staining, while lactate dehydrogenase (LDH) and creatine kinase-MB (CK-MB) levels were analyzed from coronary effluent. GSK-3ß inhibitors, SB 216763 (SB, 0.6 mg/kg, i.p.), and indirubin-3 monoxime (IND, 0.4 mg/kg, i.p.), administered 24 h before the isolation of heart, significantly decreased the I/R-induced myocardial infarct size and the release of LDH and CK-MB. The cardioprotective effect of GSK-3ß inhibitors was significantly attenuated by quercetin (4 mg/kg, i.p.), a HSP 72 inhibitor, administered 1 h before the administration of SB or IND. That the late phase of cardioprotection induced by pretreatment with GSK-3ß inhibitors is not attenuated/lost in HL rat heart is a new finding in our study. Our results indicate that HSP 72 acts on pathway of GSK-3ß and plays a significant role in cardioprotection.


Subject(s)
Glycogen Synthase Kinase 3 , HSP72 Heat-Shock Proteins , Hyperlipidemias/metabolism , Myocardium/metabolism , Animals , Diet, High-Fat , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , HSP72 Heat-Shock Proteins/antagonists & inhibitors , HSP72 Heat-Shock Proteins/metabolism , Indoles/administration & dosage , Ischemic Preconditioning, Myocardial , Male , Maleimides/administration & dosage , Myocardial Infarction/pathology , Organ Culture Techniques , Oximes/administration & dosage , Quercetin/administration & dosage , Rats , Rats, Wistar , Reperfusion Injury/metabolism , Reperfusion Injury/pathology
20.
Anticancer Res ; 32(2): 453-62, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22287732

ABSTRACT

BACKGROUND: HSP90 inhibitors effectively reduce expression and activity levels of oncogenic survival proteins. However, their clinical anti-multiple myeloma (MM) activity has been found to be rather weak, spurring the exploration of combination therapies and development of compounds with improved physicochemical properties. MATERIALS AND METHODS: Preclinical effects of the novel orally bioavailable HSP90 inhibitor NVP-HSP990 on the viability, apoptosis and client protein levels of MM cells (established cell lines and clinical specimens) were tested alone and in combination with other drugs. RESULTS: NVP-HSP990 exerted profound activity against MM cells, with a molecular mode of action conforming well with its role as HSP90 inhibitor. Enhanced activity was most obvious in combination with melphalan. Combination with a phosphatidylinositol-3-kinase (PI3-kinase)/mammalian target of rapamycin (mTOR) inhibitor, rendered the HSP90 blockade-mediated stress response ineffective and considerably increased the anti-MM toxicity. CONCLUSION: Given the current interest in both HSP90 and PI3-kinase/mTOR as potential clinical targets, these observations could broaden the therapeutic utility of either class of inhibitor in MM.


Subject(s)
HSP90 Heat-Shock Proteins/antagonists & inhibitors , Multiple Myeloma/drug therapy , Pyridones/pharmacology , Pyrimidines/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , Furans/pharmacology , HSP72 Heat-Shock Proteins/antagonists & inhibitors , Humans , Multiple Myeloma/metabolism , Phosphoinositide-3 Kinase Inhibitors , Pyridines/pharmacology , Pyridones/administration & dosage , Pyrimidines/administration & dosage , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL