Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cell Mol Biol (Noisy-le-grand) ; 68(3): 1-8, 2022 Mar 31.
Article in English | MEDLINE | ID: mdl-35988177

ABSTRACT

Anthrax is a serious infectious disease caused by Bacillus anthracis, rod-shaped gram-positive bacteria. The disease infects both humans and animals and causes severe illness.  Many vaccines have been developed for anthrax, but the vaccine with very high efficacy is yet to be developed. To overcome the problems of efficacy posed by the existing vaccines, a recombinant chimeric fusion protein containing domain 1 of lethal factor (LFD1) and domain 4 of Bacillus anthracis protective antigen (PA4) was used as antigen in copolymeric nanocapsules (NCs). Accordingly, the solvent evaporation double emulsion method was used to produce NCs containing recombinant chimeric fusion protein (LFD1-PA4). Zeta sizer and potential of nanoparticles, nanoparticle loading efficiency, release pattern of recombinant protein, and the possible effect of polylactic acid-polyethylene glycol (PLA-PEG) nanoparticle production method were investigated. Mice were used to test and evaluate the immune response. The mean titer of antibody produced against loaded LFD1-PA4 compared to free form showed a significant difference. The difference in antibody titer between the groups of once injected, twice injected, and free antigen was significant, and the highest antibody titer was found in the mice twice injected. In addition, a single-time loaded injection showed significantly higher antibodies than the free form injection indicating that loaded LFD1-PA4 into PLA-PEG nanoparticles elicits a stronger immune response. This study showed that LFD1-PA4 fusion protein from Bacillus anthracis served as an active antigen in mice. Also, the nanocarrier (PLA-PEG) containing the antigen can stimulate the immune system in the mice, owing to their controlled release property.


Subject(s)
Anthrax Vaccines , Anthrax , Bacillus anthracis , Nanocapsules , Animals , Anthrax/microbiology , Anthrax/prevention & control , Antibodies, Bacterial , Antigens, Bacterial/genetics , Bacillus anthracis/physiology , Humans , Immunity , Mice , Polyesters , Recombinant Fusion Proteins , Recombinant Proteins
2.
Mol Cell ; 81(8): 1781-1788.e4, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33571424

ABSTRACT

Mediator is a universal adaptor for transcription control. It serves as an interface between gene-specific activator or repressor proteins and the general RNA polymerase II (pol II) transcription machinery. Previous structural studies revealed a relatively small part of Mediator and none of the gene activator-binding regions. We have determined the cryo-EM structure of the Mediator at near-atomic resolution. The structure reveals almost all amino acid residues in ordered regions, including the major targets of activator proteins, the Tail module, and the Med1 subunit of the Middle module. Comparison of Mediator structures with and without pol II reveals conformational changes that propagate across the entire Mediator, from Head to Tail, coupling activator- and pol II-interacting regions.


Subject(s)
Mediator Complex Subunit 1/metabolism , Amino Acids/genetics , Protein Conformation , RNA Polymerase II/metabolism , Transcription Factors/metabolism , Transcription, Genetic/genetics
3.
Annu Rev Biochem ; 85: 715-42, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27050154

ABSTRACT

Molecular chaperones control the cellular folding, assembly, unfolding, disassembly, translocation, activation, inactivation, disaggregation, and degradation of proteins. In 1989, groundbreaking experiments demonstrated that a purified chaperone can bind and prevent the aggregation of artificially unfolded polypeptides and use ATP to dissociate and convert them into native proteins. A decade later, other chaperones were shown to use ATP hydrolysis to unfold and solubilize stable protein aggregates, leading to their native refolding. Presently, the main conserved chaperone families Hsp70, Hsp104, Hsp90, Hsp60, and small heat-shock proteins (sHsps) apparently act as unfolding nanomachines capable of converting functional alternatively folded or toxic misfolded polypeptides into harmless protease-degradable or biologically active native proteins. Being unfoldases, the chaperones can proofread three-dimensional protein structures and thus control protein quality in the cell. Understanding the mechanisms of the cellular unfoldases is central to the design of new therapies against aging, degenerative protein conformational diseases, and specific cancers.


Subject(s)
Chaperonin 60/chemistry , HSP110 Heat-Shock Proteins/chemistry , HSP70 Heat-Shock Proteins/chemistry , Heat-Shock Proteins, Small/chemistry , Mitochondrial Proteins/chemistry , Protein Unfolding , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Chaperonin 60/genetics , Chaperonin 60/metabolism , Escherichia coli/chemistry , Escherichia coli/metabolism , Gene Expression , HSP110 Heat-Shock Proteins/genetics , HSP110 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins, Small/genetics , Heat-Shock Proteins, Small/metabolism , Humans , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Models, Molecular , Protein Aggregates , Protein Folding , Protein Structure, Quaternary , Rhodospirillum rubrum/chemistry , Rhodospirillum rubrum/metabolism
4.
Cell Mol Life Sci ; 71(17): 3311-25, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24760129

ABSTRACT

By virtue of their general ability to bind (hold) translocating or unfolding polypeptides otherwise doomed to aggregate, molecular chaperones are commonly dubbed "holdases". Yet, chaperones also carry physiological functions that do not necessitate prevention of aggregation, such as altering the native states of proteins, as in the disassembly of SNARE complexes and clathrin coats. To carry such physiological functions, major members of the Hsp70, Hsp110, Hsp100, and Hsp60/CCT chaperone families act as catalytic unfolding enzymes or unfoldases that drive iterative cycles of protein binding, unfolding/pulling, and release. One unfoldase chaperone may thus successively convert many misfolded or alternatively folded polypeptide substrates into transiently unfolded intermediates, which, once released, can spontaneously refold into low-affinity native products. Whereas during stress, a large excess of non-catalytic chaperones in holding mode may optimally prevent protein aggregation, after the stress, catalytic disaggregases and unfoldases may act as nanomachines that use the energy of ATP hydrolysis to repair proteins with compromised conformations. Thus, holding and catalytic unfolding chaperones can act as primary cellular defenses against the formation of early misfolded and aggregated proteotoxic conformers in order to avert or retard the onset of degenerative protein conformational diseases.


Subject(s)
Molecular Chaperones/physiology , Protein Folding , Adenosine Triphosphate/physiology , Animals , Catalysis , Escherichia coli Proteins/physiology , Heat-Shock Proteins/physiology , Humans , Models, Biological , Molecular Chaperones/chemistry , Neurodegenerative Diseases/metabolism , Peptidylprolyl Isomerase/physiology , Protein Binding , Protein Conformation , Protein Transport , Proteostasis Deficiencies/metabolism , Stress, Physiological
5.
Front Mol Biosci ; 1: 7, 2014.
Article in English | MEDLINE | ID: mdl-25988148

ABSTRACT

The role of bacterial Hsp40, DnaJ, is to co-chaperone the binding of misfolded or alternatively folded proteins to bacterial Hsp70, DnaK, which is an ATP-fuelled unfolding chaperone. In addition to its DnaK targeting activity, DnaJ has a weak thiol-reductase activity. In between the substrate-binding domain and the J-domain anchor to DnaK, DnaJ has a unique domain with four conserved CXXC motives that bind two Zn(2+) and partly contribute to polypeptide binding. Here, we deleted in DnaJ this Zn-binding domain, which is characteristic to type I but not of type II or III J-proteins. This caused a loss of the thiol-reductase activity and strongly reduced the ability of DnaJ to mediate the ATP- and DnaK-dependent unfolding/refolding of mildly oxidized misfolded polypeptides, an inhibition that was alleviated in the presence of thioredoxin or DTT. We suggest that in addition to their general ability to target misfolded polypeptide substrates to the Hsp70/Hsp110 chaperone machinery, Type I J-proteins carry an ancillary protein dithiol-isomerase function that can synergize the unfolding action of the chaperone, in the particular case of substrates that are further stabilized by non-native disulfide bonds. Whereas the unfoldase can remain ineffective without the transient untying of disulfide bonds by the foldase, the foldase can remain ineffective without the transient ATP-fuelled unfolding of wrong local structures by the unfoldase.

6.
J Biol Chem ; 288(29): 21399-21411, 2013 Jul 19.
Article in English | MEDLINE | ID: mdl-23737532

ABSTRACT

Structurally and sequence-wise, the Hsp110s belong to a subfamily of the Hsp70 chaperones. Like the classical Hsp70s, members of the Hsp110 subfamily can bind misfolding polypeptides and hydrolyze ATP. However, they apparently act as a mere subordinate nucleotide exchange factors, regulating the ability of Hsp70 to hydrolyze ATP and convert stable protein aggregates into native proteins. Using stably misfolded and aggregated polypeptides as substrates in optimized in vitro chaperone assays, we show that the human cytosolic Hsp110s (HSPH1 and HSPH2) are bona fide chaperones on their own that collaborate with Hsp40 (DNAJA1 and DNAJB1) to hydrolyze ATP and unfold and thus convert stable misfolded polypeptides into natively refolded proteins. Moreover, equimolar Hsp70 (HSPA1A) and Hsp110 (HSPH1) formed a powerful molecular machinery that optimally reactivated stable luciferase aggregates in an ATP- and DNAJA1-dependent manner, in a disaggregation mechanism whereby the two paralogous chaperones alternatively activate the release of bound unfolded polypeptide substrates from one another, leading to native protein refolding.


Subject(s)
Adenosine Triphosphate/pharmacology , HSP110 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/metabolism , Peptides/chemistry , Peptides/metabolism , Protein Unfolding/drug effects , Biocatalysis/drug effects , Enzyme Stability/drug effects , HSP40 Heat-Shock Proteins/metabolism , Humans , Hydrolysis/drug effects , Luciferases/metabolism , Models, Biological , Protein Binding/drug effects , Protein Refolding/drug effects , Protein Stability/drug effects , Protein Structure, Quaternary , Solubility , Substrate Specificity/drug effects , Temperature , Trypsin/pharmacology
7.
Proc Natl Acad Sci U S A ; 110(18): 7199-204, 2013 Apr 30.
Article in English | MEDLINE | ID: mdl-23584019

ABSTRACT

Chaperonins are cage-like complexes in which nonnative polypeptides prone to aggregation are thought to reach their native state optimally. However, they also may use ATP to unfold stably bound misfolded polypeptides and mediate the out-of-cage native refolding of large proteins. Here, we show that even without ATP and GroES, both GroEL and the eukaryotic chaperonin containing t-complex polypeptide 1 (CCT/TRiC) can unfold stable misfolded polypeptide conformers and readily release them from the access ways to the cage. Reconciling earlier disparate experimental observations to ours, we present a comprehensive model whereby following unfolding on the upper cavity, in-cage confinement is not needed for the released intermediates to slowly reach their native state in solution. As over-sticky intermediates occasionally stall the catalytic unfoldase sites, GroES mobile loops and ATP are necessary to dissociate the inhibitory species and regenerate the unfolding activity. Thus, chaperonin rings are not obligate confining antiaggregation cages. They are polypeptide unfoldases that can iteratively convert stable off-pathway conformers into functional proteins.


Subject(s)
Adenosine Triphosphate/pharmacology , Biocatalysis/drug effects , Chaperonin 60/metabolism , Chaperonin Containing TCP-1/metabolism , Peptides/metabolism , Protein Refolding/drug effects , Protein Unfolding/drug effects , Animals , Apoproteins/metabolism , Cattle , Chaperonin 10/metabolism , Freezing , Models, Molecular , Protein Structure, Quaternary , Substrate Specificity/drug effects , Sus scrofa , Thiosulfate Sulfurtransferase/metabolism
8.
J Mol Biol ; 425(7): 1158-71, 2013 Apr 12.
Article in English | MEDLINE | ID: mdl-23306033

ABSTRACT

Misfolded polypeptide monomers may be regarded as the initial species of many protein aggregation pathways, which could accordingly serve as primary targets for molecular chaperones. It is therefore of paramount importance to study the cellular mechanisms that can prevent misfolded monomers from entering the toxic aggregation pathway and moreover rehabilitate them into active proteins. Here, we produced two stable misfolded monomers of luciferase and rhodanese, which we found to be differently processed by the Hsp70 chaperone machinery and whose conformational properties were investigated by biophysical approaches. In spite of their monomeric nature, they displayed enhanced thioflavin T fluorescence, non-native ß-sheets, and tertiary structures with surface-accessible hydrophobic patches, but differed in their conformational stability and aggregation propensity. Interestingly, minor structural differences between the two misfolded species could account for their markedly different behavior in chaperone-mediated unfolding/refolding assays. Indeed, only a single DnaK molecule was sufficient to unfold by direct clamping a misfolded luciferase monomer, while, by contrast, several DnaK molecules were necessary to unfold the more resistant misfolded rhodanese monomer by a combination of direct clamping and cooperative entropic pulling.


Subject(s)
Molecular Chaperones/chemistry , Peptides/chemistry , Protein Conformation , Protein Folding , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Biophysical Phenomena , Circular Dichroism , Electrophoresis, Polyacrylamide Gel , HSP70 Heat-Shock Proteins/chemistry , HSP70 Heat-Shock Proteins/metabolism , Kinetics , Luciferases/chemistry , Luciferases/metabolism , Models, Molecular , Molecular Chaperones/metabolism , Peptides/metabolism , Protein Multimerization , Protein Refolding , Protein Stability , Protein Structure, Secondary , Protein Unfolding , Spectroscopy, Fourier Transform Infrared , Substrate Specificity , Thiosulfate Sulfurtransferase/chemistry , Thiosulfate Sulfurtransferase/metabolism
9.
Cell Stress Chaperones ; 16(1): 15-31, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20694844

ABSTRACT

Molecular chaperones are central to cellular protein homeostasis. In mammals, protein misfolding diseases and aging cause inflammation and progressive tissue loss, in correlation with the accumulation of toxic protein aggregates and the defective expression of chaperone genes. Bacteria and non-diseased, non-aged eukaryotic cells effectively respond to heat shock by inducing the accumulation of heat-shock proteins (HSPs), many of which molecular chaperones involved in protein homeostasis, in reducing stress damages and promoting cellular recovery and thermotolerance. We performed a meta-analysis of published microarray data and compared expression profiles of HSP genes from mammalian and plant cells in response to heat or isothermal treatments with drugs. The differences and overlaps between HSP and chaperone genes were analyzed, and expression patterns were clustered and organized in a network. HSPs and chaperones only partly overlapped. Heat-shock induced a subset of chaperones primarily targeted to the cytoplasm and organelles but not to the endoplasmic reticulum, which organized into a network with a central core of Hsp90s, Hsp70s, and sHSPs. Heat was best mimicked by isothermal treatments with Hsp90 inhibitors, whereas less toxic drugs, some of which non-steroidal anti-inflammatory drugs, weakly expressed different subsets of Hsp chaperones. This type of analysis may uncover new HSP-inducing drugs to improve protein homeostasis in misfolding and aging diseases.


Subject(s)
Arabidopsis/genetics , Heat-Shock Proteins/genetics , Molecular Chaperones/genetics , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arabidopsis/metabolism , Cell Line , Computational Biology , Gene Regulatory Networks , Heat-Shock Proteins/metabolism , Hot Temperature , Humans , Molecular Chaperones/metabolism , Monocytes/metabolism , Unfolded Protein Response/genetics , Up-Regulation
10.
J Biol Chem ; 285(49): 38173-82, 2010 Dec 03.
Article in English | MEDLINE | ID: mdl-20847048

ABSTRACT

α-Synuclein aggregation and accumulation in Lewy bodies are implicated in progressive loss of dopaminergic neurons in Parkinson disease and related disorders. In neurons, the Hsp70s and their Hsp40-like J-domain co-chaperones are the only known components of chaperone network that can use ATP to convert cytotoxic protein aggregates into harmless natively refolded polypeptides. Here we developed a protocol for preparing a homogeneous population of highly stable ß-sheet enriched toroid-shaped α-Syn oligomers with a diameter typical of toxic pore-forming oligomers. These oligomers were partially resistant to in vitro unfolding by the bacterial Hsp70 chaperone system (DnaK, DnaJ, GrpE). Moreover, both bacterial and human Hsp70/Hsp40 unfolding/refolding activities of model chaperone substrates were strongly inhibited by the oligomers but, remarkably, not by unstructured α-Syn monomers even in large excess. The oligomers acted as a specific competitive inhibitor of the J-domain co-chaperones, indicating that J-domain co-chaperones may preferably bind to exposed bulky misfolded structures in misfolded proteins and, thus, complement Hsp70s that bind to extended segments. Together, our findings suggest that inhibition of the Hsp70/Hsp40 chaperone system by α-Syn oligomers may contribute to the disruption of protein homeostasis in dopaminergic neurons, leading to apoptosis and tissue loss in Parkinson disease and related neurodegenerative diseases.


Subject(s)
Bacterial Proteins/chemistry , HSP40 Heat-Shock Proteins/chemistry , HSP70 Heat-Shock Proteins/chemistry , Protein Folding , Protein Multimerization , alpha-Synuclein/chemistry , Animals , Apoptosis , Bacterial Proteins/metabolism , Cattle , HSP40 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Homeostasis , Humans , Leuconostoc/chemistry , Leuconostoc/metabolism , Lewy Bodies/chemistry , Lewy Bodies/metabolism , Neurons/metabolism , Parkinson Disease/metabolism , Protein Structure, Tertiary , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...