Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
J Cancer Res Ther ; 18(5): 1328-1337, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36204880

ABSTRACT

Aims: Alpha B-crystallin (CRYAB), a known molecular chaperone, is involved in the occurrence and development of various tumor types. However, the function of CRYAB in colorectal cancer stem cells (CSCs) remains unknown. This study aimed to elucidate the role and possible regulatory mechanisms of CRYAB in the cancer stem cell-like phenotype of colorectal cancer (CRC). Subjects and Methods: The expression of CRYAB in patients with CRC and lymph node metastasis at various stages and its relationship with overall survival were detected using the TCGA database. In this study, CRC-CSCs were enriched from HCT116 and Caco2 cells with serum-free suspension culture. The CRYAB gene, stemness-related genes, and mesenchymal markers were detected via quantitative real-time PCR (qRT-PCR) in CRC cells. Then, CRYAB-HCT116S and CRYAB-Caco2S cell lines were established by lentivirus-mediated overexpression of CRYAB. Self-renewal ability and stemness features were measured by the sphere formation assay and flow cytometry. The tumorigenesis capacity in vivo was inspected in nude mice. The functions of CRYAB on CSC proliferation, migration, and invasion were examined using colony formation and the transwell assay. Finally, the Wnt/ß-catenin pathway-related mRNAs and proteins were detected via qRT-PCR and western blotting. Results: The expression of CRYAB in CRC is related to the clinical phase and prognosis, except with lymphoid metastasis. CRYAB expression was elevated in CSCs. Upregulation of CRYAB enhanced the expression of CSC-related genes and mesenchymal markers. The capacity to form colonospheres, tumorigenesis, cell proliferation, and metastasis were significantly advanced in CRYAB-overexpressed cells. Moreover, CRYAB dramatically suppressed ß-catenin degradation and downregulated the expression of p-GSK-3ß. Conclusions: CRYAB maintains CSC formation via the Wnt/ß-catenin pathway in CRCs, which may, therefore, function as vital molecular targets for CRC therapeutic strategies.


Subject(s)
Colorectal Neoplasms , beta Catenin , Animals , Caco-2 Cells , Carcinogenesis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Glycogen Synthase Kinase 3 beta/genetics , Humans , Mice , Mice, Nude , Neoplastic Stem Cells/metabolism , Wnt Signaling Pathway/genetics , alpha-Crystallin B Chain/genetics , alpha-Crystallin B Chain/metabolism , alpha-Crystallin B Chain/pharmacology , beta Catenin/metabolism
2.
Int J Mol Sci ; 23(3)2022 Jan 20.
Article in English | MEDLINE | ID: mdl-35163023

ABSTRACT

Smaller oligomeric chaperones of α-crystallins (αA- and αB-) have received increasing attention due to their improved therapeutic potential in preventing protein aggregating diseases. Our previous study suggested that deleting 54-61 residues from the N-terminal domain (NTD) of αB-crystallin (αBΔ54-61) decreases the oligomer size and increases the chaperone function. Several studies have also suggested that NTD plays a significant role in protein oligomerization and chaperone function. The current study was undertaken to assess the effect of deleting conserved 21-28 residues from the activated αBΔ54-61 (to get αBΔ21-28, Δ54-61) on the structure-function of recombinant αBΔ21-28, Δ54-61. The αBΔ21-28, Δ54-61 mutant shows an 80% reduction in oligomer size and 3- to 25-fold increases in chaperone activity against model substrates when compared to αB-WT. Additionally, the αB∆21-28, ∆54-61 was found to prevent ß-amyloid (Aß1-42) fibril formation in vitro and suppressed Aß1-42-induced cytotoxicity in ARPE-19 cells in a more effective manner than seen with αB-WT or αB∆54-61. Cytotoxicity and reactive oxygen species (ROS) detection studies with sodium iodate (SI) showed that the double mutant protein has higher anti-apoptotic and anti-oxidative activities than the wild-type or αB∆54-61 in oxidatively stressed cells. Our study shows that the residues 21-28 and 54-61 in αB-crystallin contribute to the oligomerization and modulate chaperone function. The deletion of conserved 21-28 residues further potentiates the activated αBΔ54-61. We propose that increased substrate affinity, altered subunit structure, and assembly leading to smaller oligomers could be the causative factors for the increased chaperone activity of αBΔ21-28, Δ54-61.


Subject(s)
Antioxidants/pharmacology , Molecular Chaperones/pharmacology , Mutation , Oxidative Stress , Retinal Pigment Epithelium/drug effects , alpha-Crystallin B Chain/pharmacology , Amino Acid Sequence , Apoptosis , Cells, Cultured , Humans , Mutagenesis, Site-Directed , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , alpha-Crystallin B Chain/chemistry , alpha-Crystallin B Chain/genetics
3.
BMC Biotechnol ; 21(1): 39, 2021 06 14.
Article in English | MEDLINE | ID: mdl-34126963

ABSTRACT

BACKGROUND: HSPB5 is an ATP-independent molecular chaperone that is induced by heat shock or other proteotoxic stresses. HSPB5 is cytoprotective against stress both intracellularly and extracellularly. It acts as a potential therapeutic candidate in ischemia-reperfusion and neurodegenerative diseases. RESULTS: In this paper, we constructed a recombinant plasmid that expresses and extracellularly secrets a HSPB5-Fc fusion protein (sHSPB5-Fc) at 0.42 µg/ml in CHO-K1 cells. This sHSPB5-Fc protein contains a Fc-tag at the C-terminal extension of HSPB5, facilitating protein-affinity purification. Our study shows that sHSPB5-Fc inhibits heat-induced aggregation of citrate synthase in a time and dose dependent manner in vitro. Administration of sHSPB5-Fc protects lens epithelial cells against cisplatin- or UVB-induced cell apoptosis. It also decreases GFP-Httex1-Q74 insolubility, and reduces the size and cytotoxicity of GFP-Httex1-Q74 aggregates in PC-12 cells. CONCLUSION: This recombinant sHSPB5-Fc exhibits chaperone activity to protect cells against proteotoxicity.


Subject(s)
Protective Agents/pharmacology , alpha-Crystallin B Chain/genetics , alpha-Crystallin B Chain/pharmacology , Animals , Apoptosis/drug effects , CHO Cells , Cricetinae , Cricetulus , Cytoprotection , Epithelial Cells/chemistry , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Protective Agents/chemistry , Protective Agents/metabolism , Protein Aggregates , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , alpha-Crystallin B Chain/chemistry , alpha-Crystallin B Chain/metabolism
4.
J Thromb Thrombolysis ; 51(2): 265-276, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32621152

ABSTRACT

Acute myocardial infarction (AMI) is a life-threatening disease that often results in heart failure. CRYAB, a small heat shock protein, has been shown to have cardioprotective effects against oxidative stress-induced apoptosis in AMI. Previously, we purified a peptide derived from CRYAB (LEDQFFGEH), which we named PDFC. In this study, we determined the function of PDFC on HL-1 cardiomyocytes and explored the mechanism underlying its function. A hypoxic myocardiocyte cell line was generated by stimulation of HL-1 mouse cardiac muscle cells with different concentrations of CoCl2. Then, the hypoxic HL-1 cells were treated with the synthetic PDFC peptide, and cell proliferation, migration, and apoptosis were assessed to examine the effects of PDFC on HL-1 and hypoxic HL-1 cells. To examine the mechanism underlying the effects of PDFC on hypoxic cells, PDFC-treated hypoxic HL-1 cells were submitted for deep RNA sequencing. Finally, several differentially expressed genes in different pathways were selected for confirmation by RT-qPCR. Hypoxic myocardiocytes were generated by stimulating HL-1 cells with 800 µM CoCl2 for 24 h, which significantly upregulated HIF-1α. PDFC at 200 µg/ml showed the most positive effects on cell viability. Although hypoxic HL-1 cells and PDFC-treated hypoxic HL-1 cells both showed lower viability and migration and higher levels of apoptosis than untreated HL-1 cells, compared to hypoxic HL-1 cells, PDFC-treated hypoxic HL-1 cells showed higher viability and migration and lower apoptosis. The deep sequencing showed that 812 genes were upregulated and 1946 genes were downregulated. Among these differentially expressed genes, 699 of the upregulated genes and 1488 of the downregulated genes were protein-coding genes. Gene ontology and pathway enrichment analysis showed that the downregulated genes were dominant and that the PI3K-Akt pathway was located in the center of the network. A protein-protein interaction network was constructed, and 892 nodes were determined. In PDFC-treated hypoxic HL-1 cells, Fn1, Pik3r5, and Creb5 were downregulated, while Insr, Bcl2, Mapk14, and Pten were upregulated when compared to the levels in hypoxic HL-1 cells. In conclusion, this study reveals the significant bioactive effect of the CRYAB-derived peptide, PDFC on cardiomyocytes and the underlying mechanism.


Subject(s)
Cell Hypoxia/drug effects , Myocytes, Cardiac/drug effects , Peptides/pharmacology , Transcriptome/drug effects , alpha-Crystallin B Chain/pharmacology , Animals , Apoptosis/drug effects , Cell Line , Cobalt/adverse effects , Gene Expression Regulation/drug effects , Mice , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Peptides/chemistry , alpha-Crystallin B Chain/chemistry
5.
Int J Mol Sci ; 21(20)2020 Oct 16.
Article in English | MEDLINE | ID: mdl-33081200

ABSTRACT

Given the ability of molecular chaperones and chaperone-like proteins to inhibit the formation of pathological amyloid fibrils, the chaperone-based therapy of amyloidosis has recently been proposed. However, since these diseases are often diagnosed at the stages when a large amount of amyloids is already accumulated in the patient's body, in this work we pay attention to the undeservedly poorly studied problem of chaperone and chaperone-like proteins' effect on mature amyloid fibrils. We showed that a heat shock protein alpha-B-crystallin, which is capable of inhibiting fibrillogenesis and is found in large quantities as a part of amyloid plaques, can induce degradation of mature amyloids by two different mechanisms. Under physiological conditions, alpha-B-crystallin induces fluffing and unweaving of amyloid fibrils, which leads to a partial decrease in their structural ordering without lowering their stability and can increase their cytotoxicity. We found a higher correlation between the rate and effectiveness of amyloids degradation with the size of fibrils clusters rather than with amino acid sequence of amyloidogenic protein. Some external effects (such as an increase in medium acidity) can lead to a change in the mechanism of fibrils degradation induced by alpha-B-crystallin: amyloid fibers are fragmented without changing their secondary structure and properties. According to recent data, fibrils cutting can lead to the generation of seeds for new bona fide amyloid fibrils and accelerate the accumulation of amyloids, as well as enhance the ability of fibrils to disrupt membranes and to reduce cell viability. Our results emphasize the need to test the chaperone effect not only on fibrillogenesis, but also on the mature amyloid fibrils, including stress conditions, in order to avoid undesirable disease progression during chaperone-based therapy.


Subject(s)
Amyloid/chemistry , alpha-Crystallin B Chain/chemistry , Amyloid/drug effects , HeLa Cells , Humans , Muramidase/chemistry , Protein Conformation , alpha-Crystallin B Chain/pharmacology , beta 2-Microglobulin/chemistry
6.
Circ Heart Fail ; 10(3): e003626, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28242778

ABSTRACT

BACKGROUND: Cardiomyocytes with a less distensible titin and interstitial collagen contribute to the high diastolic stiffness of failing myocardium. Their relative contributions and mechanisms underlying loss of titin distensibility were assessed in failing human hearts. METHODS AND RESULTS: Left ventricular tissue was procured in patients with aortic stenosis (AS, n=9) and dilated cardiomyopathy (DCM, n=6). Explanted donor hearts (n=8) served as controls. Stretches were performed in myocardial strips, and an extraction protocol differentiated between passive tension (Fpassive) attributable to cardiomyocytes or to collagen. Fpassive-cardiomyocytes was higher in AS and DCM at shorter muscle lengths, whereas Fpassive-collagen was higher in AS at longer muscle lengths and in DCM at shorter and longer muscle lengths. Cardiomyocytes were stretched to investigate titin distensibility. Cardiomyocytes were incubated with alkaline phosphatase, subsequently reassessed after a period of prestretch and finally treated with the heat shock protein α-B crystallin. Alkaline phosphatase shifted the Fpassive-sarcomere length relation upward only in donor. Prestretch shifted the Fpassive-sarcomere length relation further upward in donor and upward in AS and DCM. α-B crystallin shifted the Fpassive-sarcomere length relation downward to baseline in donor and to lower than baseline in AS and DCM. In failing myocardium, confocal laser microscopy revealed α-B crystallin in subsarcolemmal aggresomes. CONCLUSIONS: High cardiomyocyte stiffness contributed to stiffness of failing human myocardium because of reduced titin distensibility. The latter resulted from an absent stiffness-lowering effect of baseline phosphorylation and from titin aggregation. High cardiomyocyte stiffness was corrected by α-B crystallin probably through relief of titin aggregation.


Subject(s)
Aortic Valve Stenosis/complications , Cardiomyopathy, Dilated/complications , Diastole/drug effects , Heart Failure/etiology , Myocytes, Cardiac/drug effects , alpha-Crystallin B Chain/pharmacology , Aged , Alkaline Phosphatase/pharmacology , Aortic Valve Stenosis/metabolism , Aortic Valve Stenosis/pathology , Aortic Valve Stenosis/physiopathology , Biomechanical Phenomena , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Dilated/physiopathology , Case-Control Studies , Collagen/metabolism , Connectin/metabolism , Female , Fluorescent Antibody Technique , Heart Failure/metabolism , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Male , Microscopy, Confocal , Middle Aged , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phosphorylation , Protein Aggregates
7.
Glia ; 65(3): 460-473, 2017 03.
Article in English | MEDLINE | ID: mdl-28063173

ABSTRACT

The glial stress protein alpha B-crystallin (HSPB5) is an endogenous agonist for Toll-like receptor 2 in CD14+ cells. Following systemic administration, HSPB5 acts as a potent inhibitor of neuroinflammation in animal models and reduces lesion development in multiple sclerosis patients. Here, we show that systemically administered HSPB5 rapidly crosses the blood-brain barrier, implicating microglia as additional targets for HSPB5 along with peripheral monocytes and macrophages. To compare key players in the HSPB5-induced protective response of human macrophages and microglia, we applied weighted gene co-expression network analysis on transcript expression data obtained 1 and 4 h after activation. This approach identified networks of genes that are co-expressed in all datasets, thus reducing the complexity of the nonsynchronous waves of transcripts that appear after activation by HSPB5. In both cell types, HSPB5 activates a network of highly connected genes that appear to be functionally equivalent and consistent with the therapeutic effects of HSPB5 in vivo, since both networks include factors that suppress apoptosis, the production of proinflammatory factors, and the development of adaptive immunity. Yet, hub genes at the core of the network in either cell type were strikingly different. They prominently feature the well-known tolerance-promoting programmed-death ligand 1 as a key player in the macrophage response to HSPB5, and the immune-regulatory enzyme cyclooxygenase-2 (COX-2) in that of microglia. This latter finding indicates that despite its reputation as a potential target for nonsteroidal anti-inflammatory drugs, microglial COX-2 plays a central role in the therapeutic effects of HSPB5 during neuroinflammation. GLIA 2017;65:460-473.


Subject(s)
Cyclooxygenase 2/metabolism , Macrophages/drug effects , Macrophages/metabolism , Microglia/drug effects , Microglia/metabolism , alpha-Crystallin B Chain/pharmacology , Animals , Brain/cytology , Cells, Cultured , Cytokines/metabolism , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Gene Regulatory Networks/drug effects , Gene Regulatory Networks/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Parenchymal Tissue/cytology , Parenchymal Tissue/drug effects , RNA, Messenger/metabolism , Time Factors , Tumor Necrosis Factor-alpha/metabolism , alpha-Crystallin B Chain/metabolism
8.
Mol Med Rep ; 12(1): 357-62, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25760062

ABSTRACT

Our group has previously shown that αB-crystallin (HSPB5), a small heat shock protein, inhibits human platelet aggregation by ristocetin, an activator of glycoprotein Ib/IX/V. In addition, it was demonstrated that glycoprotein Ib/IX/V activation induces soluble CD40 (sCD40) ligand release via thromboxane (TX) A2. In the present study, the effect of αB-crystallin on the ristocetin-induced sCD40 ligand release in human platelets was investigated. The ristocetin-induced release of sCD40 ligand was suppressed by αB-crystallin. In addition, αB-crystallin reduced the ristocetin-stimulated production of 11-dehydro-TX B2, a stable metabolite of TXA2. αB-crystallin did not suppress the platelet aggregation induced by U46619, a TXA2 receptor agonist. αB-crystallin had little effect on the U46619-induced phosphorylation of p38 mitogen-activated protein kinase or sCD40 ligand release. In addition, αB-crystallin failed to reduce the binding of SZ2, a monoclonal antibody against the sulfated sequence in the α-chain of glycoprotein Ib, to the ristocetin-stimulated platelets. These results strongly suggest that αB-crystallin extracellularly suppresses ristocetin-stimulated release of sCD40 ligand by inhibiting the TXA2 production in human platelets.


Subject(s)
Anti-Bacterial Agents/toxicity , Blood Platelets/drug effects , CD40 Ligand/metabolism , Ristocetin/toxicity , Thromboxane A2/metabolism , alpha-Crystallin B Chain/pharmacology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/toxicity , Adult , Antibodies, Monoclonal/immunology , Blood Platelets/cytology , Blood Platelets/metabolism , CD40 Ligand/analysis , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged , Phosphorylation/drug effects , Platelet Aggregation/drug effects , Thromboxane A2/analysis , alpha-Crystallin B Chain/immunology , p38 Mitogen-Activated Protein Kinases/metabolism
9.
J Control Release ; 191: 4-14, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-24780268

ABSTRACT

αB-Crystallin is a protein chaperone with anti-apoptotic and anti-inflammatory activity that is apically secreted in exosomes by polarized human retinal pigment epithelium. A 20 amino acid mini-peptide derived from residues 73-92 of αB-crystallin protects human retinal pigment epithelial (RPE) cells from oxidative stress, a process involved in the progression of age-related macular degeneration (AMD). Unfortunately, due to its small size, its development as a therapeutic requires a robust controlled release system. To achieve this goal, the αB-crystallin peptide was re-engineered into a protein polymer nanoparticle/macromolecule with the purpose of increasing the hydrodynamic radius/molecular weight and enhancing potency via multivalency or an extended retention time. The peptide was recombinantly fused with two high molecular weight (~40kDa) protein polymers inspired by human tropoelastin. These elastin-like polypeptides (ELPs) include the following: (i) a soluble peptide called S96 and (ii) a diblock copolymer called SI that assembles multivalent nanoparticles at physiological temperature. Fusion proteins, cryS96 and crySI, were found to reduce aggregation of alcohol dehydrogenase and insulin, which demonstrates that ELP fusion did not diminish chaperone activity. Next their interaction with RPE cells was evaluated under oxidative stress. Unexpectedly, H2O2-induced stress dramatically enhanced cellular uptake and nuclear localization of both cryS96 and crySI ELPs. Accompanying uptake, both fusion proteins protected RPE cells from apoptosis, as indicated by reduced caspase 3 activation and TUNEL staining. This study demonstrates the in vitro feasibility of modulating the hydrodynamic radius for small peptide chaperones by seamless fusion with protein polymers; furthermore, they may have therapeutic applications in diseases associated with oxidative stress, such as AMD.


Subject(s)
Apoptosis/drug effects , Drug Carriers , Epithelial Cells/drug effects , Molecular Chaperones , Nanoparticles , Peptide Fragments/pharmacology , Protein Engineering , Retinal Pigment Epithelium/drug effects , Technology, Pharmaceutical/methods , Tropoelastin/metabolism , alpha-Crystallin B Chain/pharmacology , Active Transport, Cell Nucleus , Caspase 3/metabolism , Cells, Cultured , Chemistry, Pharmaceutical , Cytoprotection , Delayed-Action Preparations , Enzyme Activation , Epithelial Cells/metabolism , Epithelial Cells/pathology , Feasibility Studies , Humans , Nanomedicine , Oxidative Stress , Particle Size , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Recombinant Fusion Proteins/pharmacology , Retinal Pigment Epithelium/pathology , Time Factors , Tropoelastin/chemistry , Tropoelastin/genetics , alpha-Crystallin B Chain/chemistry , alpha-Crystallin B Chain/genetics , alpha-Crystallin B Chain/metabolism
10.
Biochim Biophys Acta ; 1843(2): 309-15, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24275510

ABSTRACT

In addition to their key role as structural lens proteins, α-crystallins also appear to confer protection against many eye diseases, including cataract, retinitis pigmentosa, and macular degeneration. Exogenous recombinant α-crystallin proteins were examined for their ability to prevent cell death induced by heat or oxidative stress in a human lens epithelial cell line (HLE-B3). Wild type αA- or αB-crystallin (WT-αA and WT-αB) and αA- or αB-crystallins, modified by the addition of a cell penetration peptide (CPP) designed to enhance the uptake of proteins into cells (gC-αB, TAT-αB, gC-αA), were produced by recombinant methods. In vitro chaperone-like assays were used to assay the ability of α-crystallins to protect client proteins from chemical or heat induced aggregation. In vivo viability assays were performed in HLE-B3 to determine whether pre-treatment with α-crystallins reduced death after exposure to oxidative or heat stress. Most of the five recombinant α-crystallin proteins tested conferred some in vitro protection from protein aggregation, with the greatest effect seen with WT-αB and gC-αB. All α-crystallins displayed significant protection to oxidative stress induced cell death, while only the αB-crystallins reduced cell death induced by thermal stress. Our findings indicate that the addition of the gC tag enhanced the protective effect of αB-crystallin against oxidative but not thermally-induced cell death. In conclusion, modifications that increase the uptake of α-crystallin proteins into cells, without destroying their chaperone-like activity and anti-apoptotic functions, create the potential to use these proteins therapeutically.


Subject(s)
Crystallins/pharmacology , Cytoprotection/drug effects , Hot Temperature , Lens, Crystalline/pathology , Oxidative Stress/drug effects , alpha-Crystallin B Chain/pharmacology , Aldehyde Reductase/metabolism , Cell Death/drug effects , Cell Line , Cell Survival/drug effects , Cell-Penetrating Peptides/pharmacology , Crystallins/metabolism , Humans , Protein Structure, Quaternary , Recombinant Proteins/pharmacology , Recombinant Proteins/toxicity
11.
J Neuropathol Exp Neurol ; 72(10): 970-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24042199

ABSTRACT

Microglial nodules are frequently observed in the normal-appearing white matter of multiple sclerosis (MS) patients. Previously, we have shown that these clusters, which we call "preactive MS lesions," are closely associated with stressed oligodendrocytes and myelin sheaths that contain markedly elevated levels of the small stress protein alpha-B-crystallin (HspB5). Here, we show that microglia in these lesions express the recently identified receptors for HspB5, that is, CD14, Toll-like receptor family 1 and 2 (TLR1 and TLR2), and several molecular markers of the microglial response to HspB5. These markers were identified by genome-wide transcript profiling of 12 primary human microglial cultures at 2 time points after exposure to HspB5. These data indicate that HspB5 activates production by microglia of an array of chemokines, immune-regulatory mediators, and a striking number of antiviral genes that are generally inducible by type I interferons. Together, our data suggest that preactive MS lesions are at least in part driven by HspB5 derived from stressed oligodendrocytes and may reflect a local attempt to restore tissue homeostasis.


Subject(s)
Brain/drug effects , Microglia/drug effects , Multiple Sclerosis/metabolism , Nerve Fibers, Myelinated/drug effects , alpha-Crystallin B Chain/pharmacology , Aged , Aged, 80 and over , Axons/drug effects , Axons/metabolism , Axons/pathology , Brain/metabolism , Brain/pathology , Female , Humans , Lipopolysaccharide Receptors/genetics , Lipopolysaccharide Receptors/metabolism , Male , Microglia/immunology , Microglia/metabolism , Microglia/pathology , Middle Aged , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Myelinated/pathology , Toll-Like Receptor 1/genetics , Toll-Like Receptor 1/metabolism , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , alpha-Crystallin B Chain/metabolism
12.
J Neuroimmunol ; 259(1-2): 66-74, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23602713

ABSTRACT

CRYAB, a small heat shock protein, was previously shown to decrease neuroinflammation in experimental allergic encephalomyelitis (EAE). We investigated whether the expression of cell adhesion molecules and chemokine receptors on peripheral and spinal cord T cells, that could possibly affect their migration to the central nervous system, was altered following EAE CRYAB treatment. Less LFA-1+ lymphocytes and lower levels of iTAC, MCP-5 and MIG were observed in spinal cords of CRYAB-injected EAE animals. In addition, fewer blood T cells expressed CCR6, CXCR4 and CCR7 and in vivo-derived CRYAB EAE CD4+ lymphocytes were less migratory towards a MIP-3alpha gradient in vitro.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Cell Movement/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , alpha-Crystallin B Chain/pharmacology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Movement/immunology , Cells, Cultured , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Lymph Nodes/cytology , Mice , Mice, 129 Strain , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Neuroimmunomodulation/drug effects , Neuroimmunomodulation/immunology , Receptors, CCR6/metabolism , Receptors, CCR7/metabolism , Receptors, CXCR4/metabolism , Spinal Cord/immunology , Spinal Cord/pathology , Spleen/cytology , alpha-Crystallin B Chain/genetics , alpha-Crystallin B Chain/immunology
13.
J Biol Chem ; 287(43): 36423-34, 2012 Oct 19.
Article in English | MEDLINE | ID: mdl-22955287

ABSTRACT

To determine whether the therapeutic activity of αB crystallin, small heat shock protein B5 (HspB5), was shared with other human sHsps, a set of seven human family members, a mutant of HspB5 G120 known to exhibit reduced chaperone activity, and a mycobacterial sHsp were expressed and purified from bacteria. Each of the recombinant proteins was shown to be a functional chaperone, capable of inhibiting aggregation of denatured insulin with varying efficiency. When injected into mice at the peak of disease, they were all effective in reducing the paralysis in experimental autoimmune encephalomyelitis. Additional structure activity correlations between chaperone activity and therapeutic function were established when linear regions within HspB5 were examined. A single region, corresponding to residues 73-92 of HspB5, forms amyloid fibrils, exhibited chaperone activity, and was an effective therapeutic for encephalomyelitis. The linkage of the three activities was further established by demonstrating individual substitutions of critical hydrophobic amino acids in the peptide resulted in the loss of all of the functions.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , Paralysis/prevention & control , alpha-Crystallin B Chain/pharmacology , Amino Acid Substitution , Animals , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Mice , Mutation, Missense , Paralysis/genetics , Paralysis/metabolism , Paralysis/pathology , alpha-Crystallin B Chain/genetics
14.
PLoS One ; 7(7): e40486, 2012.
Article in English | MEDLINE | ID: mdl-22815750

ABSTRACT

As a small stress response protein, human αB crystallin, detects protein destabilization that can alter structure and function to cause self assembly of fibrils or aggregates in diseases of aging. The sensitivity of αB crystallin to protein instability was evaluated using wild-type hemoglobin (HbA) and hemoglobin S (HbS), the glutamate-6-valine mutant that forms elongated, filamentous aggregates in sickling red blood cells. The progressive thermal unfolding and aggregation of HbA and HbS in solution at 37°C, 50°C and 55°C was measured as increased light scattering. UV circular dichroism (UVCD) was used to evaluate conformational changes in HbA and HbS with time at the selected temperatures. The changes in interactions between αB crystallin and HbA or HbS with temperature were analyzed using differential centrifugation and SDS PAGE at 37°C, 50°C and 55°C. After only 5 minutes at the selected temperatures, differences in the aggregation or conformation of HbA and HbS were not observed, but αB crystallin bound approximately 6% and 25% more HbS than HbA at 37°C, and 50°C respectively. The results confirmed (a) the remarkable sensitivity of αB crystallin to structural instabilities at the very earliest stages of thermal unfolding and (b) an ability to distinguish the self assembling mutant form of HbS from the wild type HbA in solution.


Subject(s)
Hemoglobin A/chemistry , Hemoglobin A/metabolism , Hemoglobin, Sickle/chemistry , Hemoglobin, Sickle/metabolism , alpha-Crystallin B Chain/metabolism , Humans , Models, Molecular , Protein Binding , Protein Multimerization , Protein Stability/drug effects , Protein Structure, Quaternary , Protein Structure, Secondary , Protein Unfolding/drug effects , Temperature , alpha-Crystallin B Chain/pharmacology
15.
Biochemistry ; 51(12): 2588-96, 2012 Mar 27.
Article in English | MEDLINE | ID: mdl-22394327

ABSTRACT

Although a number of γD-crystallin mutations are associated with cataract formation, there is not a clear understanding of the molecular mechanism(s) that lead to this protein deposition disease. As part of our ongoing studies on crystallins, we investigated the recently discovered Arg76 to Ser (R76S) mutation that is correlated with childhood cataract in an Indian family. We expressed the R76S γD-crystallin protein in E. coli, characterized it by CD, fluorescence, and NMR spectroscopy, and determined its stability with respect to thermal and chemical denaturation. Surprisingly, no significant biochemical or biophysical differences were observed between the wild-type protein and the R76S variant, except a lowered pI (6.8 compared to the wild-type value of 7.4). NMR assessment of the R76S γD-crystallin solution structure, by RDCs, and of its motional properties, by relaxation measurements, also revealed a close resemblance to wild-type crystallin. Further, kinetic unfolding/refolding experiments for R76S and wild-type protein showed similar degrees of off-pathway aggregation suppression by αB-crystallin. Overall, our results suggest that neither structural nor stability changes in the protein are responsible for the R76S γD-crystallin variant's association with cataract. However, the change in pI and the associated surface charge or the altered nature of the amino acid could influence interactions with other lens protein species.


Subject(s)
Cataract/genetics , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mutation , gamma-Crystallins/chemistry , gamma-Crystallins/metabolism , Child , Humans , Kinetics , Models, Molecular , Mutant Proteins/genetics , Protein Multimerization/drug effects , Protein Stability/drug effects , Protein Structure, Quaternary/drug effects , Protein Unfolding/drug effects , Solubility , Thermodynamics , alpha-Crystallin B Chain/pharmacology , gamma-Crystallins/genetics
16.
J Biol Chem ; 287(13): 9708-9721, 2012 Mar 23.
Article in English | MEDLINE | ID: mdl-22308023

ABSTRACT

The therapeutic benefit of the small heat shock protein αB-crystallin (HspB5) in animal models of multiple sclerosis and ischemia is proposed to arise from its increased capacity to bind proinflammatory proteins at the elevated temperatures within inflammatory foci. By mass spectral analysis, a common set of ∼70 ligands was precipitated by HspB5 from plasma from patients with multiple sclerosis, rheumatoid arthritis, and amyloidosis and mice with experimental allergic encephalomyelitis. These proteins were distinguished from other precipitated molecules because they were enriched in the precipitate as compared with their plasma concentrations, and they exhibited temperature-dependent binding. More than half of these ligands were acute phase proteins or members of the complement or coagulation cascades. Consistent with this proposal, plasma levels of HspB5 were increased in patients with multiple sclerosis as compared with normal individuals. The combination of the thermal sensitivity of the HspB5 combined with the high local concentration of these ligands at the site of inflammation is proposed to explain the paradox of how a protein believed to exhibit nonspecific binding can bind with some relative apparent selectivity to proinflammatory proteins and thereby modulate inflammation.


Subject(s)
Blood Proteins/immunology , Molecular Chaperones/pharmacology , Multiple Sclerosis/blood , alpha-Crystallin B Chain/pharmacology , Amyloidosis/blood , Amyloidosis/drug therapy , Animals , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/drug therapy , Encephalomyelitis, Autoimmune, Experimental , Female , Humans , Inflammation/blood , Inflammation/drug therapy , Mice , Mice, Inbred BALB C , Molecular Chaperones/blood , Multiple Sclerosis/drug therapy , Protein Binding , alpha-Crystallin B Chain/blood
17.
Proc Natl Acad Sci U S A ; 108(32): 13287-92, 2011 Aug 09.
Article in English | MEDLINE | ID: mdl-21828004

ABSTRACT

Tissue plasminogen activator is the only treatment option for stroke victims; however, it has to be administered within 4.5 h after symptom onset, making its use very limited. This report describes a unique target for effective treatment of stroke, even 12 h after onset, by the administration of αB-crystallin (Cryab), an endogenous immunomodulatory neuroprotectant. In Cryab(-/-) mice, there was increased lesion size and diminished neurologic function after stroke compared with wild-type mice. Increased plasma Cryab was detected after experimental stroke in mice and after stroke in human patients. Administration of Cryab even 12 h after experimental stroke reduced both stroke volume and inflammatory cytokines associated with stroke pathology. Cryab is an endogenous anti-inflammatory and neuroprotectant molecule produced after stroke, whose beneficial properties can be augmented when administered therapeutically after stroke.


Subject(s)
Stroke/drug therapy , Stroke/immunology , alpha-Crystallin B Chain/therapeutic use , Animals , Brain/drug effects , Brain/immunology , Brain/pathology , Humans , Immune System/drug effects , Immune System/immunology , Mice , Stroke/blood , Stroke/pathology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Time Factors , alpha-Crystallin B Chain/administration & dosage , alpha-Crystallin B Chain/blood , alpha-Crystallin B Chain/pharmacology
18.
Ann Thorac Surg ; 91(6): 1907-13, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21619989

ABSTRACT

BACKGROUND: This study investigates the protective effect of exogenous αB-crystallin (CryAB) on myocardial function after ischemia-reperfusion injury. METHODS: Mice underwent temporary left anterior descending artery occlusion for 30 minutes. Either CryAB (50 µg) or phosphate-buffered saline (100 µL [n=6, each group]) were injected in the intramyocardial medial and lateral perinfarct zone 15 minutes before reperfusion. Intraperitoneal injections were administered every other day. Left ventricular ejection fraction was evaluated on postoperative day 40 with magnetic resonance imaging. To investigate the effect of CryAB on apoptosis after hypoxia/reoxygenation in vitro, murine atrial cardiomyocytes (HL-1 cells) or human microvascular endothelial cells (HMEC-1) were incubated with either 50 µg CryAB (500 µg /10 mL) or phosphate-buffered saline in a hypoxia chamber for 6, 12, and 24 hours, followed by 30 minutes of reoxygenation at room air. Apoptosis was then assessed by western blot (Bcl-2, free bax, cleaved caspases-3, 9, PARP) and enzyme-linked immunosorbent assay analyses (cytoplasmic histone-associated DNA fragments and caspase-3 activity). RESULTS: On postoperative day 40, CryAB-treated mice had a 1.8-fold increase in left ventricular ejection fraction versus control mice (27%±6% versus 15%±4% SD, p<0.005). In vitro, (1) the HL-1 cells showed no significant difference in apoptotic protein expression, cytoplasmic histone-associated DNA fragments, or caspase-3 activity; (2) the HMEC-1 cells had increased but not significant apoptotic protein expression with, however, a significant decrease in cytoplasmic histone-associated DNA fragments (1.5-fold, p<0.01) and caspase-3 activity (2.7-fold, p<0.005). CONCLUSIONS: Exogenous CryAB administration significantly improves cardiac function after ischemia-reperfusion injury, in vivo. The protective anti-apoptotic affects of CryAB may target the endothelial cell.


Subject(s)
Apoptosis/drug effects , Endothelial Cells/drug effects , Myocardial Reperfusion Injury/drug therapy , Ventricular Function, Left/drug effects , alpha-Crystallin B Chain/pharmacology , Animals , Cell Line , Humans , Male , Mice , Mice, Inbred C57BL , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology
19.
J Cell Biochem ; 112(9): 2454-62, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21538481

ABSTRACT

Cell-penetrating peptides (CPPs), including TAT-CPP, have been used to deliver exogenous proteins into living cells. Although a number of proteins fused to TAT-CPP can be delivered into various cells, little is known about the proteolytic cleavage of TAT-fusion proteins in cells. In this study, we demonstrate that a small heat shock protein (sHSP), alphaB-crystallin (αB-crystallin), delivered by TAT-CPP is susceptible to proteolytic cleavage by matrix metalloproteinase-1 (MMP-1) in cardiac myoblast H9c2 cells. Recombinant TAT-αB-crystallin was efficiently transduced into H9c2 cells. For a few hours following protein transduction, generation of a 14-kDa fragment, a cleavage band of TAT-αB-crystallin, increased in a time-dependent manner. This fragment was observed only in detergent-insoluble fractions. Interestingly, treatment with MMP inhibitors blocked the cleavage of TAT-αB-crystallin. In test tubes, recombinant MMP-1 processed TAT-αB-crystallin to generate the major cleavage fragment 14-kDa, as observed in the cells treated with TAT-αB-crystallin. The N-terminal sequences of the 14-kDa fragment were identified as Leu-Arg-Ala-Pro-Ser-Trp-Phe, indicating that this fragment is generated by cleavage at Phe54-Leu55 of αB-crystallin. The MMP-1-selective inhibitor abolished the production of 14-kDa fragments in cells. In addition, the cleaved fragment of TAT-αB-crystallin was significantly reduced in cells transfected with MMP-1 siRNA. Moreover, the enzymatic activity of MMP-1 was markedly increased in TAT-αB-crystallin-treated cells. TAT-αB-crystallin has a cytoprotective effect on H9c2 cells under hypoxic insult, moreover, MMP-1-selective inhibitor treatment led to even increased cell viability. These results suggest that MMP-1 is responsible for proteolytic cleavage of TAT-αB-crystallin during its intracellular transduction in H9c2 cells.


Subject(s)
Cell-Penetrating Peptides/pharmacokinetics , Matrix Metalloproteinase 1/metabolism , Recombinant Fusion Proteins/pharmacokinetics , alpha-Crystallin B Chain/pharmacokinetics , tat Gene Products, Human Immunodeficiency Virus/pharmacokinetics , Animals , Cell Hypoxia , Cell Line , Cell Survival/drug effects , Cell-Penetrating Peptides/isolation & purification , Cell-Penetrating Peptides/pharmacology , Cytoprotection , Drug Delivery Systems , Enzyme Assays , Matrix Metalloproteinase 1/genetics , Myoblasts/drug effects , Myoblasts/enzymology , Myoblasts/metabolism , Rats , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/pharmacology , alpha-Crystallin B Chain/isolation & purification , alpha-Crystallin B Chain/pharmacology , tat Gene Products, Human Immunodeficiency Virus/isolation & purification , tat Gene Products, Human Immunodeficiency Virus/pharmacology
20.
FEBS Lett ; 583(15): 2464-8, 2009 Aug 06.
Article in English | MEDLINE | ID: mdl-19559024

ABSTRACT

alphaB-crystallin, a low-molecular-weight heat shock protein (HSP), has binding sites on platelets. However, the exact role of alphaB-crystallin is not clarified. In this study, we investigated the effect of alphaB-crystallin on platelet granule secretion. alphaB-crystallin attenuated the adenosine diphosphate (ADP)-induced phosphorylation of p44/p42 mitogen-activated protein kinase (MAPK) and p38 MAPK. The ADP-stimulated HSP27 phosphorylation was markedly reduced by alphaB-crystallin. alphaB-crystallin significantly suppressed the ADP-induced secretions of both platelet-derived growth factor (PDGF)-AB and serotonin. Therefore, our results strongly suggest that alphaB-crystallin extracellularly suppresses platelet granule secretion by inhibition of HSP27 phosphorylation via p44/p42 MAPK and p38 MAPK.


Subject(s)
Adenosine Diphosphate/metabolism , Blood Platelets/drug effects , Blood Platelets/metabolism , Secretory Vesicles/metabolism , alpha-Crystallin B Chain/pharmacology , Blood Platelets/cytology , HSP27 Heat-Shock Proteins/metabolism , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...