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










Database
Language
Publication year range
1.
Photochem Photobiol Sci ; 18(2): 546-554, 2019 Feb 13.
Article in English | MEDLINE | ID: mdl-30681107

ABSTRACT

Photodynamic therapy (PDT) is a clinically approved treatment that exerts a selective cytotoxic activity toward cancer cells. The procedure involves the administration of a photosensitizer drug followed by its activation by visible light. In the presence of oxygen, a series of events lead to tumor cell death. PDT releases different cell signals, some of these lead to death while others can lead to survival. The surviving or resistant cells contribute to the recurrence of tumors after treatment, from which the necessity to understand this molecular response induced by PDT arises. It has been shown that both Heat Shock Proteins (HSPs) and autophagy promote PDT resistance. Moreover, both of them can be stimulated by PDT treatment. However, the molecular interplay between HSPs and autophagy in the photodynamic therapy context is poorly understood. We studied whether PDT induces autophagic activity through HSPs. We demonstrated that PDT promoted HSP27 expression, which in turn triggered autophagic cell survival as well as inhibited apoptosis in colon cancer cells. In addition, an overexpression of the HSP27/autophagy axis was observed in skin carcinoma cells resistant to PDT.


Subject(s)
Autophagy/drug effects , Autophagy/radiation effects , HSP27 Heat-Shock Proteins/metabolism , Photochemotherapy , Caco-2 Cells , Cell Survival/drug effects , Cell Survival/radiation effects , Gene Knockdown Techniques , HSP27 Heat-Shock Proteins/deficiency , HSP27 Heat-Shock Proteins/genetics , Humans , Photosensitizing Agents/pharmacology , Protoporphyrins/pharmacology
2.
Cardiovasc Res ; 115(1): 154-167, 2019 01 01.
Article in English | MEDLINE | ID: mdl-29982352

ABSTRACT

Aims: Inadequate healing after myocardial infarction (MI) leads to heart failure and fatal ventricular rupture, while optimal healing requires timely induction and resolution of inflammation. This study tested the hypothesis that heat shock protein B1 (HSPB1), which limits myocardial inflammation during endotoxemia, modulates wound healing after MI. Methods and results: To test this hypothesis, cardiomyocyte-specific HSPB1 knockout (Hspb1-/-) mice were generated using the Cre-LoxP recombination system. MI was induced by ligation of the left anterior descending coronary artery in Hspb1-/- and wild-type (WT) littermates. HSPB1 was up-regulated in cardiomyocytes of WT animals in response to MI, and deficiency of cardiomyocyte HSPB1 increased MI-induced cardiac rupture and mortality within 21 days after MI. Serial echocardiography showed more aggravated remodelling and cardiac dysfunction in Hspb1-/- mice than in WT mice at 1, 3, and 7 days after MI. Decreased collagen deposition and angiogenesis, as well as increased MMP2 and MMP9 activity, were also observed in Hspb1-/- mice compared with WT controls after MI, using immunofluorescence, polarized light microscopy, and zymographic analyses. Notably, Hspb1-/- hearts exhibited enhanced and prolonged leucocyte infiltration, enhanced expression of inflammatory cytokines, and enhanced TLR4/MyD88/NFκB activation compared with WT controls after MI. In-depth molecular analyses in both mice and primary cardiomyocytes demonstrated that cardiomyocyte-specific knockout of HSPB1 increased nuclear factor-κB (NFκB) activation, which promoted the expression of proinflammatory mediators. This led to increased leucocyte recruitment, thereby to excessive inflammation, ultimately resulting in adverse remodelling, cardiac dysfunction, and cardiac rupture following MI. Conclusion: These data suggest that HSPB1 acts as a negative regulator of NFκB-mediated leucocyte recruitment and the subsequent inflammation in cardiomyocytes. Cardiomyocyte HSPB1 is required for wound healing after MI and could be a target for myocardial repair in MI patients.


Subject(s)
Chemotaxis, Leukocyte , Heart Rupture, Post-Infarction/metabolism , Heat-Shock Proteins/deficiency , Leukocytes/metabolism , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , NF-kappa B/metabolism , Ventricular Remodeling , Animals , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , HSP27 Heat-Shock Proteins/deficiency , HSP27 Heat-Shock Proteins/genetics , Heart Rupture, Post-Infarction/immunology , Heart Rupture, Post-Infarction/pathology , Heart Rupture, Post-Infarction/physiopathology , Heat-Shock Proteins/genetics , Leukocytes/immunology , Mice, Inbred C57BL , Mice, Knockout , Molecular Chaperones/genetics , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/immunology , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/pathology , Neovascularization, Physiologic , Rats, Sprague-Dawley , Signal Transduction , Wound Healing
3.
J Vis Exp ; (115)2016 09 27.
Article in English | MEDLINE | ID: mdl-27768052

ABSTRACT

Small interfering RNA (siRNA) is among the most widely used RNA interference methods for the short-term silencing of protein-coding genes. siRNA is a synthetic RNA duplex created to specifically target a mRNA transcript to induce its degradation and it has been used to identify novel pathways in various cellular processes. Few reports exist regarding the role of phosphorylated heat shock protein 27 (HSP27) in corneal epithelial wound healing. Herein, cultured human corneal epithelial cells were divided into a scrambled control-siRNA transfected group and a HSP27-specific siRNA-transfected group. Scratch-induced directional wounding assays, and western blotting, and flow cytometry were then performed. We conclude that HSP27 has roles in corneal epithelial wound healing that may involve epithelial cell apoptosis and migration. Here, step-by-step descriptions of sample preparation and the study protocol are provided.


Subject(s)
Cornea/physiology , HSP27 Heat-Shock Proteins/physiology , Wound Healing/physiology , Blotting, Western , Cell Line , Cells, Cultured , Cornea/cytology , Cornea/metabolism , Epithelial Cells/metabolism , Epithelial Cells/physiology , Flow Cytometry , Gene Knockdown Techniques/methods , HSP27 Heat-Shock Proteins/deficiency , HSP27 Heat-Shock Proteins/genetics , HSP27 Heat-Shock Proteins/metabolism , Heat-Shock Proteins , Humans , Molecular Chaperones , Phosphorylation , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Transfection/methods
4.
Cancer Res ; 76(5): 1019-30, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26744531

ABSTRACT

The endothelial-to-mesenchymal transition (EndMT) contributes to cancer, fibrosis, and other pathologic processes. However, the underlying mechanisms are poorly understood. Endothelial HSP1 (HSPB1) protects against cellular stress and has been implicated in cancer progression and pulmonary fibrosis. In this study, we investigated the role of HSPB1 in mediating the EndMT during the development of pulmonary fibrosis and lung cancer. HSPB1 silencing in human pulmonary endothelial cells accelerated emergence of the fibrotic phenotype after treatment with TGFß or other cytokines linked to pulmonary fibrosis, suggesting that HSPB1 maintains endothelial cell identity. In mice, endothelial-specific overexpression of HSPB1 was sufficient to inhibit pulmonary fibrosis by blocking the EndMT. Conversely, HSPB1 depletion in a mouse model of lung tumorigenesis induced the EndMT. In clinical specimens of non-small cell lung cancer, HSPB1 expression was absent from tumor endothelial cells undergoing the EndMT. Our results showed that HSPB1 regulated the EndMT in lung fibrosis and cancer, suggesting that HSPB1-targeted therapeutic strategies may be applicable for treating an array of fibrotic diseases.


Subject(s)
Epithelial-Mesenchymal Transition , HSP27 Heat-Shock Proteins/physiology , Heat-Shock Proteins/physiology , Lung Neoplasms/prevention & control , Neoplasm Proteins/physiology , Pulmonary Fibrosis/prevention & control , Animals , Cells, Cultured , Disease Models, Animal , Epithelial-Mesenchymal Transition/radiation effects , HSP27 Heat-Shock Proteins/analysis , HSP27 Heat-Shock Proteins/deficiency , Heat-Shock Proteins/analysis , Humans , Janus Kinases/physiology , Lung Neoplasms/etiology , Mice , Mice, Transgenic , Molecular Chaperones , Neoplasm Proteins/analysis , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Pulmonary Fibrosis/etiology , STAT3 Transcription Factor/physiology
5.
Methods Mol Biol ; 1364: 127-42, 2016.
Article in English | MEDLINE | ID: mdl-26472447

ABSTRACT

Small interfering RNA (SiRNA) delivery remains a major challenge in RNAi-based therapy. Dendrimers are emerging as appealing nonviral vectors for SiRNA delivery thanks to their well-defined architecture and their unique cooperativity and multivalency confined within a nanostructure. We have recently demonstrated that structurally flexible poly(amidoamine) (PAMAM) dendrimers are safe and effective nanovectors for SiRNA delivery in various disease models in vitro and in vivo. The present chapter showcases these dendrimers can package different SiRNA molecules into stable and nanosized particles, which protect SiRNA from degradation and promote cellular uptake of SiRNA, resulting in potent gene silencing at both mRNA and protein level in the prostate cancer cell model. Our results demonstrate this set of flexible PAMAM dendrimers are indeed safe and effective nonviral vectors for SiRNA delivery and hold great promise for further applications in functional genomics and RNAi-based therapies.


Subject(s)
Dendrimers/chemistry , Drug Carriers/chemistry , Nanoparticles/chemistry , RNA, Small Interfering/chemistry , Biological Transport , Cell Line, Tumor , Dendrimers/metabolism , Drug Carriers/metabolism , Drug Stability , Gene Expression Regulation/genetics , Gene Silencing , HSP27 Heat-Shock Proteins/deficiency , HSP27 Heat-Shock Proteins/genetics , Humans , Microscopy, Confocal , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Ribonuclease, Pancreatic/metabolism
6.
PLoS One ; 8(10): e77383, 2013.
Article in English | MEDLINE | ID: mdl-24143227

ABSTRACT

There is large literature describing in vitro experiments on heat shock protein (hsp)B1 but understanding of its function in vivo is limited to studies in mice overexpressing human hspB1 protein. Experiments in cells have shown that hspB1 has chaperone activity, a cytoprotective role, regulates inflammatory gene expression, and drives cell proliferation. To investigate the function of the protein in vivo we generated hspB1-deficient mice. HspB1-deficient fibroblasts display increased expression of the pro-inflammatory cytokine, interleukin-6, compared to wild-type cells, but reduced proliferation. HspB1-deficient fibroblasts exhibit reduced entry into S phase and increased expression of cyclin-dependent kinase inhibitors p27(kip1) and p21(waf1). The expression of hspB1 protein and mRNA is also controlled by the cell cycle. To investigate the physiological function of hspB1 in regulating inflammation and cell proliferation we used an excisional cutaneous wound healing model. There was a significant impairment in the rate of healing of wounds in hspB1-deficient mice, characterised by reduced re-epithelialisation and collagen deposition but also increased inflammation. HspB1 deficiency augments neutrophil infiltration in wounds, driven by increased chemokine (C-X-C motif) ligand 1 expression. This appears to be a general mechanism as similar results were obtained in the air-pouch and peritonitis models of acute inflammation.


Subject(s)
HSP27 Heat-Shock Proteins/deficiency , Wound Healing , Animals , Cell Cycle/drug effects , Cell Proliferation , Collagen/metabolism , Epithelial Cells/cytology , Epithelial Cells/drug effects , Exons/genetics , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , HSP27 Heat-Shock Proteins/genetics , Interleukin-1/pharmacology , Interleukin-6/metabolism , Mice , Peritonitis/chemically induced , Peritonitis/pathology , Peritonitis/physiopathology , Skin/cytology , Skin/injuries , Skin/metabolism , Zymosan/adverse effects
7.
J Biol Chem ; 286(38): 33289-300, 2011 Sep 23.
Article in English | MEDLINE | ID: mdl-21784846

ABSTRACT

Mutant p53 accumulation has been shown to induce the multidrug resistance gene (MDR1) and ATP binding cassette (ABC)-based drug efflux in human breast cancer cells. In the present work, we have found that transcriptional activation of the oxidative stress-responsive heat shock factor 1 (HSF-1) and expression of heat shock proteins, including Hsp27, which is normally known to augment proteasomal p53 degradation, are inhibited in Adriamycin (doxorubicin)-resistant MCF-7 cells (MCF-7/adr). Such an endogenous inhibition of HSF-1 and Hsp27 in turn results in p53 mutation with gain of function in its transcriptional activity and accumulation in MCF-7/adr. Also, lack of HSF-1 enhances nuclear factor κB (NF-κB) DNA binding activity together with mutant p53 and induces MDR1 gene and P-glycoprotein (P-gp, ABCB1), resulting in a multidrug-resistant phenotype. Ectopic expression of Hsp27, however, significantly depleted both mutant p53 and NF-κB (p65), reversed the drug resistance by inhibiting MDR1/P-gp expression in MCF-7/adr cells, and induced cell death by increased G(2)/M population and apoptosis. We conclude from these results that HSF-1 inhibition and depletion of Hsp27 is a trigger, at least in part, for the accumulation of transcriptionally active mutant p53, which can either directly or NF-κB-dependently induce an MDR1/P-gp phenotype in MCF-7 cells. Upon Hsp27 overexpression, this pathway is abrogated, and the acquired multidrug resistance is significantly abolished so that MCF-7/adr cells are sensitized to Dox. Thus, clinical alteration in Hsp27 or NF-κB level will be a potential approach to circumvent drug resistance in breast cancer.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Breast Neoplasms/genetics , Doxorubicin/metabolism , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , HSP27 Heat-Shock Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Breast Neoplasms/pathology , Cell Death/drug effects , Cell Line, Tumor , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Drug Resistance, Neoplasm/genetics , Enzyme Activation/drug effects , Female , HSP27 Heat-Shock Proteins/deficiency , Heat Shock Transcription Factors , Heat-Shock Proteins , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Molecular Chaperones , Mutant Proteins/metabolism , NF-kappa B/metabolism , Phosphorylation/drug effects , Signal Transduction/drug effects , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism
8.
PLoS One ; 6(2): e16550, 2011 Feb 17.
Article in English | MEDLINE | ID: mdl-21379584

ABSTRACT

Molecular chaperones protect cells from the deleterious effects of protein misfolding and aggregation. Neurotoxicity of amyloid-beta (Aß) aggregates and their deposition in senile plaques are hallmarks of Alzheimer's disease (AD). We observed that the overall content of αB-crystallin, a small heat shock protein molecular chaperone, decreased in AD model mice in an age-dependent manner. We hypothesized that αB-crystallin protects cells against Aß toxicity. To test this, we crossed αB-crystallin/HspB2 deficient (CRYAB⁻/⁻HSPB2⁻/⁻) mice with AD model transgenic mice expressing mutant human amyloid precursor protein. Transgenic and non-transgenic mice in chaperone-sufficient or deficient backgrounds were examined for representative behavioral paradigms for locomotion and memory network functions: (i) spatial orientation and locomotion was monitored by open field test; (ii) sequential organization and associative learning was monitored by fear conditioning; and (iii) evoked behavioral response was tested by hot plate method. Interestingly, αB-crystallin/HspB2 deficient transgenic mice were severely impaired in locomotion compared to each genetic model separately. Our results highlight a synergistic effect of combining chaperone deficiency in a transgenic mouse model for AD underscoring an important role for chaperones in protein misfolding diseases.


Subject(s)
Alzheimer Disease/complications , Alzheimer Disease/pathology , Disease Models, Animal , HSP27 Heat-Shock Proteins/genetics , Mental Disorders/etiology , Mice, Transgenic , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Animals , Avoidance Learning/physiology , Behavior, Animal/physiology , HSP27 Heat-Shock Proteins/deficiency , Humans , Locomotion/genetics , Locomotion/physiology , Male , Mental Disorders/diagnosis , Mental Disorders/genetics , Mice , Mice, Inbred C57BL , Molecular Chaperones/genetics , Pain Measurement , Physical Conditioning, Animal , alpha-Crystallin B Chain/genetics
9.
Am J Physiol Cell Physiol ; 299(2): C363-73, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20427712

ABSTRACT

Hypoxia triggers responses in endothelial cells that play roles in many conditions including high-altitude pulmonary edema and tumor angiogenesis. Signaling pathways activated by hypoxia modify cytoskeletal and contractile proteins and alter the biomechanical properties of endothelial cells. Intermediate filaments are major components of the cytoskeleton whose contribution to endothelial physiology is not well understood. We have previously shown that hypoxia-activated signaling in endothelial cells alters their contractility and adhesiveness. We have also linked p38-MAP kinase signaling pathway leading to HSP27 phosphorylation and increased actin stress fiber formation to endothelial barrier augmentation. We now show that vimentin, a major intermediate filament protein in endothelial cells, is regulated by hypoxia. Our results indicate that exposure of endothelial cells to hypoxia causes vimentin filament networks to initially redistribute perinuclearly. However, by 1 hour hypoxia these networks reform and appear more continuous across cells than under normoxia. Hypoxia also causes transient changes in vimentin phosphorylation, and activation of PAK1, a kinase that regulates vimentin filament assembly. In addition, exposure to 1 hour hypoxia increases the ratio of insoluble/soluble vimentin. Overexpression of phosphomimicking mutant HSP27 (pmHSP27) causes changes in vimentin distribution that are similar to those observed in hypoxic cells. Knocking-down HSP27 destroys the vimentin filamentous network, and disrupting vimentin filaments with acrylamide increases endothelial permeability. Both hypoxia- and pmHSP27 overexpression-induced changes are reversed by inhibition of phosphatase activity. In conclusion hypoxia causes redistribution of vimentin to a more insoluble and extensive filamentous network that could play a role in endothelial barrier stabilization. Vimentin redistribution appears to be mediated through altering the phosphorylation of the protein and its interaction with HSP27.


Subject(s)
Endothelial Cells/physiology , Intermediate Filaments/physiology , Vimentin/physiology , Animals , Cell Hypoxia/physiology , Cells, Cultured , Endothelial Cells/metabolism , Gene Knockdown Techniques , HSP27 Heat-Shock Proteins/deficiency , HSP27 Heat-Shock Proteins/genetics , Phosphorylation/physiology , Rats , Vimentin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...