Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
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
2.
Biomed Res ; 38(3): 175-182, 2017.
Article in English | MEDLINE | ID: mdl-28637952

ABSTRACT

Increase of thrombus in the coronary arteries is positively correlated with the level of heat-shock protein 72 (HSP72) in the blood of patients with acute myocardial infarction (AMI). Platelet aggregation participates in thrombus formation on ruptured plaque in AMI. In this study, we aimed to clarify the role of HSP72 in thrombus formation by evaluating the effects of HSP72 on platelet aggregation. Platelet aggregation activities were measured in platelet-rich plasma obtained from male Sprague-Dawley rats with or without the platelet activators, such as adenosine diphosphate (ADP), collagen, thrombin receptor-activating peptide-6 (TRAP-6), ristocetin, and arachidonic acid. Changes in aggregation were estimated by the co-addition of recombinant HSP72 and anti-HSP72 antibodies. Our results showed that addition of HSP72 increased platelet aggregation in the presence of low concentrations of ADP, collagen, TRAP-6, ristocetin, and arachidonic acid. Increased platelet aggregation stimulated by ADP and HSP72 was reduced by the co-addition of anti-HSP72 antibodies. Thus, these findings suggested that HSP72 was released extracellularly in response to stress, promoting thrombus formation and AMI. Additionally, treatment with anti-HSP72 antibodies may control platelet aggregation induced by extracellular HSP72.


Subject(s)
HSP72 Heat-Shock Proteins/physiology , Platelet Aggregation , Adenosine Diphosphate/physiology , Animals , Blood Coagulation Factors/physiology , Blood Platelets/physiology , Collagen/physiology , Male , Peptide Fragments/physiology , Rats, Sprague-Dawley
3.
J Nephrol ; 30(1): 11-18, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27059325

ABSTRACT

The study of kidney development at the cellular and molecular levels remains an active area of nephrology research. The functional integrity of the kidney depends on normal development as well as on physiological cell turnover. Apoptosis induction is essential for these mechanisms. A route to cell death revealed in the past decade shows that heat shock proteins (HSPs) and their cofactors are responsible for regulating the apoptotic pathway. Specifically, heat shock protein 70 (Hsp70), the most ubiquitous and highly conserved HSP, helps proteins adopt native conformation or regain function after misfolding. Hsp70 is an important cofactor for the function of Wilms' tumour 1 (WT1) and suggests a potential role for this chaperone during kidney differentiation. In addition, we have demonstrated that WT1 expression is modulated by nitric oxide (NO) availability and Hsp70 interaction after neonatal unilateral ureteral obstruction. NO has been identified as playing an important role in the developing kidney. These findings suggest that Hsp70 and NO may play a critical and fundamental role in the capacity to modulate both apoptotic pathway and oxidative stress during kidney development. Furthermore, the design of experimental protocols that assess renal epithelial functionality in this context, could contribute to the understanding of renal development and alterations.


Subject(s)
HSP70 Heat-Shock Proteins/physiology , Kidney/embryology , Nitric Oxide/physiology , WT1 Proteins/physiology , Adult , Animals , Apoptosis , Epithelial-Mesenchymal Transition , HSP72 Heat-Shock Proteins/physiology , Humans , Oxidative Stress
4.
Nat Struct Mol Biol ; 23(9): 821-9, 2016 09.
Article in English | MEDLINE | ID: mdl-27478930

ABSTRACT

Hsp70s use ATP hydrolysis to disrupt protein-protein associations and to move macromolecules. One example is the Hsc70- mediated disassembly of the clathrin coats that form on vesicles during endocytosis. Here, we exploited the exceptional features of these coats to test three models-Brownian ratchet, power-stroke and entropic pulling-proposed to explain how Hsp70s transform their substrates. Our data rule out the ratchet and power-stroke models and instead support a collision-pressure mechanism whereby collisions between clathrin-coat walls and Hsc70s drive coats apart. Collision pressure is the complement to the pulling force described in the entropic pulling model. We also found that self-association augments collision pressure, thereby allowing disassembly of clathrin lattices that have been predicted to be resistant to disassembly. These results illuminate how Hsp70s generate the forces that transform their substrates.


Subject(s)
Clathrin Heavy Chains/chemistry , HSP72 Heat-Shock Proteins/physiology , Protein Multimerization , Amino Acid Sequence , Animals , Binding Sites , Cryoelectron Microscopy , Entropy , HSP72 Heat-Shock Proteins/chemistry , Hydrogen-Ion Concentration , Models, Molecular , Particle Size , Protein Domains , Protein Stability , Protein Structure, Quaternary , Rats
5.
J Surg Res ; 198(1): 165-74, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26073348

ABSTRACT

BACKGROUND: This study was undertaken to clarify the role of extracellular heat shock protein 72 on the survival of sepsis and to determine possible factor(s) that may be responsible for it. MATERIALS AND METHODS: Sepsis was induced by cecal ligation and puncture. Changes in serum levels of heat shock protein (Hsp72) and cytokines were determined during sepsis, and the results were correlated with the survival. Effects of heat pretreatment on Hsp72 expression in septic rat leukocytes and those of septic rat serum, lipopolysaccharide (LPS), and certain cytokines on the release of Hsp72 in macrophage NR8383 cells were determined. RESULTS: Circulating Hsp72 levels were increased during the progress of sepsis (0, 5.5, 6.5, 10, and 6.5 ng/mL at 0, 3, 6, 9, and 18 h after cecal ligation and puncture, respectively) and the increases were correlated positively with survival rates. LPS triggered the release of Hsp72 in heat pretreated animals. Heat pretreatment increased Hsp72 expression in nonsepsis (+535%, P < 0.01) and sepsis (+116%, P<0.01%) rat leukocytes. Incubation of sepsis rat serum with NR8383 cells increased levels of extracellular heat shock protein 72 in cultured medium. Cytokine profiling revealed that among the 19 cytokines screened, four of them were increased as follows: cytokine-induced neutrophil chemoattractant 3 (+211.3%, P < 0.05), interleukin 10 (+147%, P < 0.05), MCP-1 (+49.6%, P < 0.05), and tumor necrosis factor alpha (+51.8%, P < 0.05). MCP-1 and LPS were capable of releasing Hsp72 from NR8383 cells. CONCLUSIONS: These results demonstrate that the increases in the levels of circulating Hsp72 had a beneficial effect in improving animal survival during the progress of sepsis. The increases in circulating Hsp72 may be mediated via MCP-1 and/or LPS.


Subject(s)
HSP72 Heat-Shock Proteins/physiology , Sepsis/mortality , Animals , Cell Line , Chemokine CCL2/physiology , Cytokines/analysis , Leukocytes/chemistry , Lipopolysaccharides/toxicity , Male , Rats , Rats, Sprague-Dawley , Sepsis/immunology
6.
Biochim Biophys Acta ; 1853(1): 52-62, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25260982

ABSTRACT

Hyperthermia (39-45°C) has emerged as an alternate prospect for cancer therapy in combination with radiation and chemotherapy. Despite promising progress in the clinic, molecular mechanisms involved in hyperthermia-induced cell death are not clear. Hyperthermia causes protein denaturation/aggregation, which results in cell death by apoptosis and/or necrosis. Hyperthermia also induces thermotolerance, which renders cells resistant to subsequent exposure to lethal heat shock. This study investigates the role of both lethal (42-43°C) and mild (40°C) hyperthermia in regulating ER stress and ER stress-induced apoptosis in HeLa cells. The ability of mild thermotolerance induced at 40°C to alleviate either or both of these processes is also determined. Hyperthermia (42-43°C) induced ER stress, revealed by phosphorylation of PERK, eIF2α and IRE1α, cleavage of ATF6 and increased expression of BiP and sXBP1. Real-time PCR revealed that mRNA levels of ATF6, ATF4, BiP, sXBP1 and CHOP increased in cells exposed to hyperthermia. Moreover, hyperthermia caused disruption of calcium homeostasis and activated the calpain-calpastatin proteolytic system and ER resident caspase 4. Pre-exposure to mild hyperthermia (40°C) alleviated the induction of cytotoxicity and ER stress by hyperthermia (42-43°C) and protected cells against ER stress-induced apoptosis. ShRNA-mediated depletion of Hsp72 abrogated protective effects of mild thermotolerance (40°C) against heat-shock induced ER stress and sensitized cells to ER stress-mediated apoptosis. Our findings show that Hsp72 contributes to the protective effects of mild hyperthermia (40°C) against hyperthermia-induced ER stress and apoptosis.


Subject(s)
Apoptosis , Endoplasmic Reticulum Stress , Heat-Shock Response , Calpain/metabolism , Caspases/metabolism , HSP72 Heat-Shock Proteins/physiology , HeLa Cells , Hot Temperature , Humans
7.
J Anim Sci ; 92(7): 3026-34, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24879765

ABSTRACT

Heat stress adversely affects the productivity and immune status of dairy cows. The temperature-humidity index (THI) is commonly used to indicate the degree of heat stress on dairy cattle. We investigated the effects of different THI and Cr supplementation on the antioxidant capacity, the levels of heat shock protein 72 (Hsp72), and cytokine responses of lactating cows. The study used a total of 24 clinically healthy uniparous midlactation Holstein cows, which were randomly divided into 2 groups (n = 12 per group), and was conducted in 3 designated THI periods: low THI period (LTHI; THI = 56.4 ± 2.5), moderate THI period (MTHI; THI = 73.9 ± 1.7), and high THI period (HTHI; THI = 80.3 ± 1.0). The 2 groups of cows were fed corn and corn silage based basal diet supplemented chromium picolinate to provide 3.5 mg of Cr/cow daily (Cr+) or basal diet with no Cr (Cr-). The experiment was a 3 × 2 factorial design. The numbers of leukocytes (P < 0.05) and serum levels of glucose (P < 0.001) were lower; however, the serum levels of blood urea nitrogen (BUN; P < 0.001) and creatinine (P < 0.001) were greater in the MTHI and HTHI than in LTHI. The total antioxidant capacity in the serum was unaltered; an increase in superoxide dismutase activity (P < 0.001) and in serum malondialdehyde concentration (P < 0.001) was observed in the MTHI and HTHI compared with the LTHI. The high THI led to increases in serum concentrations of tumor necrosis factor-α (TNF-α; P < 0.001) and IL-10 (P < 0.05). Cows supplemented with Cr had lower (P = 0.009) serum concentrations of cholesterol but greater (P < 0.001, respectively) serum levels of Hsp72 and IL-10 compared with those without Cr supplementation in the HTHI. Western blot analysis revealed that cows supplemented with Cr had greater (P = 0.038) expression of the inhibitor of nuclear factor kappa B α (IκBα) in peripheral blood mononuclear cells (PBMC) compared with those without Cr supplementation in the HTHI, whereas the expression of Hsp72 in PBMC was unaltered. Data indicate that there is a decrease in glucose and increases in BUN and creatinine in the serum of midlactation cows under hot conditions during the summer and that these cows have a lowered oxidative capacity but an elevated antioxidant capacity. In addition, Cr may play an anti-inflammatory role in lactating cows by promoting the release of Hsp72, increasing the production of IL-10, and inhibiting the degradation of IκBα under hot conditions during the summer.


Subject(s)
Cattle/physiology , Cytokines/blood , HSP72 Heat-Shock Proteins/blood , Heat-Shock Response/physiology , Humidity/adverse effects , Lactation/physiology , Oxidation-Reduction/drug effects , Picolinic Acids/pharmacology , Animals , Cattle/immunology , Cytokines/physiology , Diet/veterinary , Dietary Supplements , Female , HSP72 Heat-Shock Proteins/physiology , Heat-Shock Response/drug effects , Hot Temperature/adverse effects , Interleukin-10/blood , Lactation/drug effects , Malondialdehyde/blood , NF-kappa B/blood , Superoxide Dismutase/blood , Tumor Necrosis Factor-alpha/blood
8.
Anesthesiology ; 120(6): 1450-62, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24667831

ABSTRACT

BACKGROUND: The heat-shock response (HSR) protects from insults, such as ischemia-reperfusion injury, by inhibiting signaling pathways activated by sterile inflammation. However, the mechanisms by which the HSR activation would modulate lung damage and host response to a bacterial lung infection remain unknown. METHODS: HSR was activated with whole-body hyperthermia or by intraperitoneal geldanamycin in mice that had their lungs instilled with Pseudomonas aeruginosa 24 h later (at least six mice per experimental group). Four hours after instillation, lung endothelial and epithelial permeability, bacterial counts, protein levels in bronchoalveolar lavage fluid, and lung myeloperoxidase activity were measured. Mortality rate 24 h after P. aeruginosa instillation was recorded. The HSR effect on the release of interleukin-10 and killing of P. aeruginosa bacteria by a mouse alveolar macrophage cell line and on neutrophil phagocytosis was also examined. RESULTS: HSR activation worsened lung endothelial (42%) and epithelial permeability (50%) to protein, decreased lung bacterial clearance (71%), and increased mortality (50%) associated with P. aeruginosa pneumonia, an effect that was not observed in heat-shock protein-72-null mice. HSR-mediated decrease in neutrophil phagocytosis (69%) and bacterial killing (38%) by macrophages was interleukin-10 dependent, a mechanism confirmed by increased lung bacterial clearance and decreased mortality (70%) caused by P. aeruginosa pneumonia in heat-shocked interleukin-10-null mice. CONCLUSIONS: Prior HSR activation worsens lung injury associated with P. aeruginosa pneumonia in mice via heat-shock protein-72- and interleukin-10-dependent mechanisms. These results provide a novel mechanism for the immunosuppression observed after severe trauma that is known to activate HSR in humans.


Subject(s)
HSP72 Heat-Shock Proteins/physiology , Interleukin-10/physiology , Lung Injury/metabolism , Pseudomonas Infections/metabolism , Pseudomonas aeruginosa , Up-Regulation/immunology , Animals , Cell Line , Cells, Cultured , Heat-Shock Response/immunology , Interleukin-10/metabolism , Lung Injury/immunology , Lung Injury/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pseudomonas Infections/immunology , Random Allocation , Sheep
9.
Diabetes ; 63(3): 1124-39, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24270982

ABSTRACT

We have investigated the role of heat shock (HS) in preventing insulin resistance-induced endothelial dysfunction. To the best of our knowledge, we report here for the first time that insulin resistance inhibits vascular HS protein (HSP) 72 expression. HS treatment (41 °C for 20 min) restored the HSP72 expression. High-fat diet (HFD)-fed, insulin-resistant rats show attenuated angiotensin (ANG)-(1-7)-induced vasodilator effect, endothelial nitric oxide synthase (eNOS) phosphorylation, AMP-activated protein kinase phosphorylation, and sirtuin 1 (SIRT1) expression. Interestingly, HS prevented this attenuation. We also provide the first evidence that HFD-fed rats show increased vascular DNA methyltransferase 1 (DNMT1) expression and that HS prevented this increase. Our data show that in HFD-fed rats HS prevented loss in the expression of ANG-(1-7) receptor Mas and ACE2, which were responsible for vascular complications. Further, the inhibition of eNOS (l-N(G)-nitro-L-arginine methyl ester), Mas (A-779), and SIRT1 (nicotinamide) prevented the favorable effects of HS. This suggests that HS augmented ANG-(1-7) signaling via the Mas/eNOS/SIRT1 pathway. Our study, for the first time, suggests that induction of intracellular HSP72 alters DNMT1 expression, and may function as an epigenetic regulator of SIRT1 and eNOS expression. We propose that induction of HSP72 is a novel approach to prevent insulin resistance-induced vascular complications.


Subject(s)
Angiotensin I/pharmacology , HSP72 Heat-Shock Proteins/physiology , Insulin Resistance , Peptide Fragments/pharmacology , Signal Transduction/physiology , AMP-Activated Protein Kinases/physiology , Animals , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation , Diet, High-Fat , Hot Temperature , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/physiology , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , Sirtuin 1/physiology , Vasodilation/drug effects
10.
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
11.
J Neurol Sci ; 317(1-2): 123-9, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22386689

ABSTRACT

Although recent researches show that Heat Shock Protein 72 (HSP72) plays an important role in neuronal survival, little knowledge is known about the precise mechanisms during cerebral ischemia/reperfusion (I/R). Our present study investigated the neuroprotective mechanisms of HSP72 against ischemic brain injury induced by cerebral I/R. Mild heat shock pretreatment was employed to induce the overexpression of HSP72 by immersing rats into the water bath at 42°C for 20 min before cerebral I/R. HSP72 antisense oligodeoxynucleotides (ODNs) were used to inhibit HSP72 expression by intracerebroventricular infusion once per day for 3 days before cerebral I/R animal model was induced by four-vessel occlusion for 15 min transient ischemia and then reperfused for various time in Sprague-Dawley rats. Immunoprecipitation and immunoblotting were used to detect the expression of the related proteins. HE-staining and TUNEL-staining were carried out to examine the neuronal death of hippocampal CA1 region. Results showed that mild heat shock could increase the phosphorylation of protein kinase B (Akt), inhibit the assembly of MLK3-MKK7-JNK3 signaling module, diminish the phosphorylation of JNK3 and c-Jun, and decrease the activation of caspase-3. Furthermore, mild heat shock could significantly protect neurons against cerebral I/R. Whereas, all of the aforementioned effects of mild heat shock were reversed by HSP72 antisense ODNs. In summary, our results imply that Akt1 activation is involved in the neuroprotection of HSP72 against ischemic brain injury via suppressing JNK3 signaling pathway and provide a new experimental foundation for stroke therapy.


Subject(s)
Brain Ischemia/metabolism , HSP72 Heat-Shock Proteins/physiology , MAP Kinase Signaling System/genetics , Mitogen-Activated Protein Kinase 10/antagonists & inhibitors , Mitogen-Activated Protein Kinase 10/physiology , Proto-Oncogene Proteins c-akt/physiology , Animals , Brain Ischemia/prevention & control , HSP72 Heat-Shock Proteins/biosynthesis , Infusions, Intraventricular , Male , Oligonucleotides, Antisense/administration & dosage , Rats , Rats, Sprague-Dawley
12.
Pigment Cell Melanoma Res ; 25(1): 88-98, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21978301

ABSTRACT

HSP70i and other stress proteins have been used in anti-tumor vaccines. This begs the question whether HSP70i plays a unique role in immune activation. We vaccinated inducible HSP70i (Hsp70-1) knockout mice and wild-type animals with optimized TRP-1, a highly immunogenic melanosomal target molecule. We were unable to induce robust and lasting depigmentation in the Hsp70-1 knockout mice, and in vivo cytolytic assays revealed a lack of cytotoxic T-lymphocyte activity. Absence of T-cell infiltration to the skin and maintenance of hair follicle melanocytes were observed. By contrast, depigmentation proceeded without interruption in mice lacking a tissue-specific constitutive isoform of HSP70 (Hsp70-2) vaccinated with TRP-2. Next, we demonstrated that HSP70i was necessary and sufficient to accelerate depigmentation in vitiligo-prone Pmel-1 mice, accompanied by lasting phenotypic changes in dendritic cell subpopulations. In summary, these studies assign a unique function to HSP70i in vitiligo and identify HSP70i as a targetable entity for treatment.


Subject(s)
HSP72 Heat-Shock Proteins/physiology , Vitiligo/immunology , Animals , Autoantigens/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Dendritic Cells/pathology , Female , HSP72 Heat-Shock Proteins/deficiency , HSP72 Heat-Shock Proteins/genetics , Inflammation , Intramolecular Oxidoreductases/immunology , Male , Melanocytes/immunology , Melanocytes/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Oxidoreductases/immunology , Skin Pigmentation/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccination , Vitiligo/pathology , gp100 Melanoma Antigen/immunology
13.
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
15.
Cell Stress Chaperones ; 16(4): 379-87, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21188661

ABSTRACT

The present studies aimed to understand the interrelationships between stress, hormones and heat shock proteins (HSPs) in the ovary. We examined (1) whether HSP70.2, HSP72 and HSP105/110 can be produced and accumulated in porcine ovarian tissue, (2) whether these HSPs could be indicators of stress, i.e. whether two kinds of stress (high temperatures and malnutrition/serum deprivation) can affect them, and (3) whether some hormonal regulators of ovarian functions (insulin-like growth factor (IGF)-I, leptin and follicle-stimulating hormone (FSH)) can affect these HSPs and response of ovaries to HSP-related stress. We analysed the expression of HSP70.2, HSP72 and HSP105/110 mRNA (by using real-time reverse transcriptase polymerase chain reaction) in porcine ovarian granulosa cells, as well as the accumulation of HSP70 protein (by using sodium dodecyl sulphate polyacrylamide gel electrophoresis-Western) in either whole ovarian follicles and granulose cells cultured at normal (37.5°C) or high (41.5°C) temperature, with and without serum and with and without IGF-I, leptin and FSH. Expression of mRNA for HSP70.2, HSP72 and HSP105/110 in ovarian granulosa cells and accumulation of HSP70 protein in whole ovarian follicles and granulosa cells were demonstrated. In all the groups, addition of either IGF-I, leptin and FSH reduced the expression of HSP70.2, HSP72 and HSP105/110 mRNA. Both high temperature, serum deprivation and their combination resulted in increase in mRNAs for all three analysed HSPs. Additions of either IGF-I, leptin and FSH prevented the stimulatory effect of both high temperature and serum deprivation on the transcription of HSP70.2, HSP72 and HSP105/110. In contrast, high temperature reduced accumulation of peptide HSP70 in both ovarian follicles and granulosa cell. Serum deprivation promoted accumulation of HSP70 in granulosa cells, but not in ovarian follicles. Addition of IGF-I, leptin and FSH was able to alter accumulation of HSP70 in both follicles and granulosa cells. The present observations suggest (1) that HSPs can be synthesised in ovarian follicular granulosa cells; (2) that hormones (IGF-I, leptin and FSH) can inhibit, whilst stressors (both high temperature and malnutrition/serum deprivation) can stimulate transcription of HSP70.2, HSP72 and HSP105/110 genes, whilst heat stress, but not malnutrition, can promote depletion of HSP70 in ovarian cells, and (3) that hormones (IGF-I, leptin and FSH) can prevent stress-related changes in HSPs. The application of HSPs as indicators and mediators of stress and hormones on ovarian functions, as well as use of hormones and HSPs as anti-stressor molecules, are discussed.


Subject(s)
Follicle Stimulating Hormone/metabolism , Heat-Shock Proteins/metabolism , Insulin-Like Growth Factor I/metabolism , Leptin/metabolism , Ovary/metabolism , Stress, Physiological , Animals , Female , Food Deprivation/physiology , Granulosa Cells/metabolism , Granulosa Cells/physiology , HSP110 Heat-Shock Proteins/metabolism , HSP110 Heat-Shock Proteins/physiology , HSP72 Heat-Shock Proteins/metabolism , HSP72 Heat-Shock Proteins/physiology , Heat-Shock Proteins/biosynthesis , Ovarian Follicle/metabolism , Ovarian Follicle/physiology , Ovary/physiology , Swine , Temperature
16.
Hypertens Res ; 33(10): 975-80, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20686485

ABSTRACT

Primary (essential) hypertension has been shown to be mediated by a relative impairment in sodium excretion by the kidney, but the mechanisms responsible for this defect are still being clarified. Increasing evidence suggests a role for subtle acquired renal injury in mediating this process. Microvascular injury is present in the majority of subjects with hypertension. The development of arteriolosclerosis, primarily of the afferent arteriole, may interfere with glomerular autoregulation, whereas the loss of peritubular capillaries may facilitate local ischemia. These changes favor the localization of T cells and macrophages into the interstitium, which, coupled with local oxidative stress and angiotensin II generation, may contribute to the impaired pressure natriuresis observed with salt-sensitive hypertension. Consistent with this hypothesis, therapies that are aimed at blocking the immune response, including thymectomy, genetic alterations in mice resulting in impaired immune responses, or the use of immunosuppressive agents, can protect against the development of hypertension in experimental models. Preliminary data in humans also suggest that the inhibition of the renal inflammatory response may reduce blood pressure. The present investigations are directed to gain insight in the role of the intrarenal T-cell reactivity and autoimmunity in driving the tubulointerstitial inflammation and its participation in the pathogenesis of salt-sensitive hypertension.


Subject(s)
Hypertension/physiopathology , Kidney Diseases/physiopathology , Nephritis, Interstitial/physiopathology , Animals , HSP72 Heat-Shock Proteins/physiology , Humans , Mice , Microvessels/physiopathology , Models, Animal , Natriuresis/physiology , Oxidative Stress/physiology
17.
Radiother Oncol ; 95(3): 350-8, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20430459

ABSTRACT

BACKGROUND AND PURPOSE: Hsp72 found in the extracellular milieu has been shown to play an important role in immune regulation. The impact of common cancer therapies on extracellular release of Hsp72 however, has been to date undefined. MATERIALS AND METHODS: Serum from 13 patients undergoing radiation therapy (XRT) for prostate cancer with or without hormonal therapy (ADT) was measured for levels of circulating serum Hsp72 and pro-inflammatory cytokines (IL-6 and TNF-alpha) using the classical sandwich ELISA technique and the relative expression of CD8(+) T lymphocytes and natural killer (NK) cells was measured using flow cytometry. Mouse orthotopic xenograft of human prostate cancer tumors (DU-145 and PC-3) were used to validate and further characterize the response noted in the clinical setting. The biological significance of tumor released Hsp72 was studied in human dendritic cells (DC) in vitro. RESULTS: Circulating serum Hsp72 levels increased an average of 3.5-fold (median per patient 4.8-fold) with XRT but not with ADT (p=0.0002). Increases in IL-6 (3.3-fold), TNF-alpha (1.8-fold), CD8(+) CTL (2.1-fold) and NK cells (3.2-fold) also occurred. Using PC-3 and DU-145 human prostate cancer xenograft models in mice, we confirmed that XRT induces Hsp72 release primarily from implanted tumors. In vitro studies using supernatant recovered from irradiated human prostate cancer cells point to exosomes containing Hsp72 as a possible stimulator of pro-inflammatory cytokine production and costimulatory molecules expression in human DC. CONCLUSIONS: The current study confirms for the first time in an actual clinical setting elevation of circulating serum Hsp72 with XRT. The accompanying studies in mice and in vitro identify the released exosomes containing Hsp72 as playing a pivotal role in stimulating pro-inflammatory immune responses. These findings, if validated, may lead to new treatment paradigms for common human malignancies.


Subject(s)
HSP72 Heat-Shock Proteins/blood , Prostatic Neoplasms/radiotherapy , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Exosomes/metabolism , HSP27 Heat-Shock Proteins/blood , HSP72 Heat-Shock Proteins/physiology , Humans , Killer Cells, Natural/immunology , Male , Mice , Mice, Inbred BALB C , Prostatic Neoplasms/blood , Prostatic Neoplasms/immunology , Tumor Necrosis Factor-alpha/blood , Xenograft Model Antitumor Assays
18.
J Am Soc Nephrol ; 21(4): 598-609, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20133478

ABSTRACT

Although heat shock protein 72 (HSP72) ameliorates renal tubulointerstitial fibrosis by inhibiting epithelial-to-mesenchymal transition (EMT), the underlying mechanism is unknown. Because Smad proteins transduce TGF-beta signaling from the cytosol to the nucleus and HSP72 assists in protein folding and facilitates nuclear translocation, we investigated whether HSP72 inhibits TGF-beta-induced EMT by modulating Smad expression, activation, and nuclear translocation. To evaluate the roles of distinct HSP72 structural domains in these processes, we constructed vectors that expressed wild-type HSP72 or mutants lacking either the peptide-binding domain (HSP72-DeltaPBD), which is responsible for substrate binding and refolding, or the nuclear localization signal (HSP72-DeltaNLS). Overexpression of wild-type HSP72 or HSP72-DeltaNLS inhibited TGF-beta1-induced EMT, but HSP72-DeltaPBD did not, suggesting a critical role for the PBD in this inhibition. HSP72 overexpression inhibited TGF-beta1-induced phosphorylation and nuclear translocation of Smad3 and p-Smad3, but not Smad2; these inhibitory effects required the PBD but not the NLS. Coimmunoprecipitation assays suggested a physical interaction between Smad3 and the PBD. siRNA knockdown of endogenous HSP72 enhanced both TGF-beta1-induced Smad3 phosphorylation and EMT and confirmed the interaction of HSP72 with both Smad3 and p-Smad3. In vivo, induction of HSP72 by geranylgeranylacetone suppressed Smad3 phosphorylation in renal tubular cells after unilateral ureteral obstruction. In conclusion, HSP72 inhibits EMT in renal epithelial cells primarily by exerting domain-specific effects on Smad3 activation and nuclear translocation.


Subject(s)
Epithelial Cells/cytology , HSP72 Heat-Shock Proteins/physiology , Mesoderm/cytology , Smad3 Protein/physiology , Urothelium/cytology , Animals , Cell Differentiation , Kidney , Male , Mice , Protein Transport , Rats , Rats, Sprague-Dawley
19.
J Clin Invest ; 120(2): 457-71, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20093776

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) have been identified in humans and mice as a population of immature myeloid cells with the ability to suppress T cell activation. They accumulate in tumor-bearing mice and humans and have been shown to contribute to cancer development. Here, we have isolated tumor-derived exosomes (TDEs) from mouse cell lines and shown that an interaction between TDE-associated Hsp72 and MDSCs determines the suppressive activity of the MDSCs via activation of Stat3. In addition, tumor-derived soluble factors triggered MDSC expansion via activation of Erk. TDE-associated Hsp72 triggered Stat3 activation in MDSCs in a TLR2/MyD88-dependent manner through autocrine production of IL-6. Importantly, decreasing exosome production using dimethyl amiloride enhanced the in vivo antitumor efficacy of the chemotherapeutic drug cyclophosphamide in 3 different mouse tumor models. We also demonstrated that this mechanism is relevant in cancer patients, as TDEs from a human tumor cell line activated human MDSCs and triggered their suppressive function in an Hsp72/TLR2-dependent manner. Further, MDSCs from cancer patients treated with amiloride, a drug used to treat high blood pressure that also inhibits exosome formation, exhibited reduced suppressor functions. Collectively, our findings show in both mice and humans that Hsp72 expressed at the surface of TDEs restrains tumor immune surveillance by promoting MDSC suppressive functions.


Subject(s)
HSP72 Heat-Shock Proteins/physiology , Amiloride/pharmacology , Amiloride/therapeutic use , Animals , Cell Line , Cell Line, Tumor , Cyclophosphamide/therapeutic use , Exosomes/drug effects , Exosomes/immunology , Exosomes/physiology , Humans , Immunosuppression Therapy , Mice , Mice, Nude , Neoplasms/drug therapy , Neoplasms/immunology , T-Lymphocytes/immunology , T-Lymphocytes, Regulatory/immunology
20.
Int J Hyperthermia ; 25(8): 626-33, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20021223

ABSTRACT

PURPOSE: Hyperthermia-induced activation of stress response proteins allows cells to withstand metabolic insults. In this study we set out to determine whether insulin secretion by pancreatic beta cells was affected by the acute inflammatory response, systemic inflammation-induced hyperglycaemia, and whole-body hyperthermia. Given that systemic-inflammation induces ER stress, we further examined whether hyperthermia can attenuate the extent of LPS-induced ER stress. MATERIALS AND METHODS: Rats were randomised and divided into three treatment groups. Control rats received a 0.9% NaCl solution. Rats in the lipopolysaccharide (LPS) group received 7.5 mg of LPS/kg. Rats in the whole-body hyperthermia (WBH) + LPS group were exposed to 42 degrees C for 15 min, followed by injection with 7.5 mg of LPS/kg after 48 h. Glucose-potentiated insulin release and extent of ER stress were measured in beta cells. RESULTS: LPS inhibited glucose-induced insulin release from islet cells and induced the expression of Bip/GRP78, XBP-1, and CHOP transcripts. The inhibition of glucose-induced insulin release and induction of ER stress proteins by LPS was attenuated by WBH. CONCLUSIONS: Our findings suggest that LPS-induced systemic inflammation decreased insulin release due to the effects of ER stress proteins on insulin secretion. Furthermore, the induction of ER stress proteins was prevented by pretreating rats with WBH. This may suggest that inhibiting the induction of ER stress proteins through WBH can restore insulin release in various disease states.


Subject(s)
Endoplasmic Reticulum/drug effects , HSP72 Heat-Shock Proteins/physiology , Insulin-Secreting Cells/physiology , Animals , Fever/metabolism , Hyperthermia, Induced , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Lipopolysaccharides/pharmacology , Male , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...