Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 119(14): e2113520119, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35349341

ABSTRACT

SignificanceClassic serine proteases are synthesized as inactive precursors that are proteolytically processed, resulting in irreversible activation. We report an alternative and reversible mechanism of activation that is executed by an inactive protease. This mechanism involves a protein complex between the serine protease HTRA1 and the cysteine protease calpain 2. Surprisingly, activation is restricted as it improves the proteolysis of soluble tau protein but not the dissociation and degradation of its amyloid fibrils, a task that free HTRA1 is efficiently performing. These data exemplify a challenge for protein quality control proteases in the clearing of pathogenic fibrils and suggest a potential for unexpected side effects of chemical modulators targeting PDZ or other domains located at a distance to the active site.


Subject(s)
Calpain , Serine Endopeptidases , Amyloid/metabolism , Calpain/metabolism , High-Temperature Requirement A Serine Peptidase 1/chemistry , Proteolysis , Serine Endopeptidases/metabolism , Serine Proteases/metabolism
2.
Biol Chem ; 402(11): 1427-1440, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34472763

ABSTRACT

Glycosaminoglycans (GAGs) are essential functional components of the extracellular matrix (ECM). Artificial GAGs like sulfated hyaluronan (sHA) exhibit pro-osteogenic properties and boost healing processes. Hence, they are of high interest for supporting bone regeneration and wound healing. Although sulfated GAGs (sGAGs) appear intracellularly, the knowledge about intracellular effects and putative interaction partners is scarce. Here we used an affinity-purification mass spectrometry-based (AP-MS) approach to identify novel and particularly intracellular sGAG-interacting proteins in human bone marrow stromal cells (hBMSC). Overall, 477 proteins were found interacting with at least one of four distinct sGAGs. Enrichment analysis for protein localization showed that mainly intracellular and cell-associated interacting proteins were identified. The interaction of sGAG with α2-macroglobulin receptor-associated protein (LRPAP1), exportin-1 (XPO1), and serine protease HTRA1 (HTRA1) was confirmed in reverse assays. Consecutive pathway and cluster analysis led to the identification of biological processes, namely processes involving binding and processing of nucleic acids, LRP1-dependent endocytosis, and exosome formation. Respecting the preferentially intracellular localization of sGAG in vesicle-like structures, also the interaction data indicate sGAG-specific modulation of vesicle-based transport processes. By identifying many sGAG-specific interacting proteins, our data provide a resource for upcoming studies aimed at molecular mechanisms and understanding of sGAG cellular effects.


Subject(s)
Glycosaminoglycans/metabolism , High-Temperature Requirement A Serine Peptidase 1/metabolism , Karyopherins/metabolism , LDL-Receptor Related Protein-Associated Protein/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Cells, Cultured , Chromatography, Liquid , Glycosaminoglycans/chemistry , High-Temperature Requirement A Serine Peptidase 1/chemistry , High-Temperature Requirement A Serine Peptidase 1/isolation & purification , Humans , Karyopherins/chemistry , Karyopherins/isolation & purification , LDL-Receptor Related Protein-Associated Protein/chemistry , LDL-Receptor Related Protein-Associated Protein/isolation & purification , Mesenchymal Stem Cells/chemistry , Mesenchymal Stem Cells/metabolism , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/isolation & purification , Tandem Mass Spectrometry , Exportin 1 Protein
3.
ACS Chem Biol ; 15(9): 2346-2354, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32786264

ABSTRACT

The high temperature requirement A (HTRA) family of serine proteases mediates protein quality control. These proteins process misfolded proteins in several diseases including Alzheimer's disease (AD) and Parkinson's disease (PD). While their structures and activation mechanisms have been studied, the precise details of the regulation of their activity under physiological conditions have not been completely elucidated, partly due to the lack of suitable chemical probes. In the present study, we developed novel activity-based probes (ABPs) targeting the HTRAs and demonstrated their utility in the monitoring and quantification of changes in enzyme activity in live cells. Using our probes, we found the activity of HTRA1 to be highly elevated in an AD-like cell-based model. We also observed the active HTRA2 in live cells by using a mitochondrion-targeted probe. We believe that our probes can serve as a useful tool to study the role of human HTRAs in neurodegenerative diseases.


Subject(s)
Fluoresceins/chemistry , Fluorescent Dyes/chemistry , High-Temperature Requirement A Serine Peptidase 1/metabolism , High-Temperature Requirement A Serine Peptidase 2/metabolism , Molecular Probes/chemistry , Organophosphonates/chemistry , Cell Line, Tumor , High-Temperature Requirement A Serine Peptidase 1/chemistry , High-Temperature Requirement A Serine Peptidase 2/chemistry , Humans , Microscopy, Confocal , Microscopy, Fluorescence , Mitochondria/metabolism , Oligopeptides/chemistry
4.
Mol Genet Genomic Med ; 8(6): e1111, 2020 06.
Article in English | MEDLINE | ID: mdl-32239807

ABSTRACT

BACKGROUND: We investigated whether a heterozygous mutation that we newly identified in HTRA1 (high-temperature requirement serine protease A1 gene) in a pedigree with autosomal dominant hereditary cerebral small vessel disease (SVD) reduces the function of HTRA1 and affects the transforming growth factor-ß1 (TGF-ß1)/Smad signaling. METHODS: Whole-exome sequence from the proband and her two sisters was examined using whole-exome enrichment and sequencing. Expression of HTRA1 and TGF-ß1/Smad and HTRA1 activity were assayed using sodium dodecyl sulfate-polyacrylamide gel electrophoresis and western blotting analyses after transfecting wild-type and mutant HTRA1 genes into HEK293 cells. RESULTS: A new heterozygous mutation (c.614C>G:p.Ser205Cys) in HTRA1 was identified in the sequence encoding the trypsin-like serine protease domain. The mutation was predicted to be deleterious by in silico tools. Moreover, in vitro activity and protein analyses revealed a loss-of-function effect of the mutation: the proteolytic activity of mutant HTRA1 was decreased, and, notably, this was accompanied by an increase in TGF-ß1/Smad protein levels. CONCLUSIONS: The heterozygous mutation HTRA1 S205C causing diminished protease activity is associated with-and could represent a cause of-autosomal dominant hereditary cerebral SVD. Our results also indicate a relationship between HTRA1 and TGF-ß1/Smad signaling.


Subject(s)
Cerebral Small Vessel Diseases/genetics , High-Temperature Requirement A Serine Peptidase 1/genetics , Mutation , Cerebral Small Vessel Diseases/pathology , Female , Genes, Dominant , HEK293 Cells , High-Temperature Requirement A Serine Peptidase 1/chemistry , High-Temperature Requirement A Serine Peptidase 1/metabolism , Humans , Middle Aged , Protein Domains , Signal Transduction , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism
5.
Proc Natl Acad Sci U S A ; 117(3): 1414-1418, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31907318

ABSTRACT

Startling reports described the paradoxical triggering of the human mitogen-activated protein kinase pathway when a small-molecule inhibitor specifically inactivates the BRAF V600E protein kinase but not wt-BRAF. We performed a conceptual analysis of the general phenomenon "activation by inhibition" using bacterial and human HtrA proteases as models. Our data suggest a clear explanation that is based on the classic biochemical principles of allostery and cooperativity. Although substoichiometric occupancy of inhibitor binding sites results in partial inhibition, this effect is overrun by a concomitant activation of unliganded binding sites. Therefore, when an inhibitor of a cooperative enzyme does not reach saturating levels, a common scenario during drug administration, it may cause the contrary of the desired effect. The implications for drug development are discussed.


Subject(s)
Allosteric Site , Antineoplastic Agents/pharmacology , Heat-Shock Proteins/antagonists & inhibitors , High-Temperature Requirement A Serine Peptidase 1/antagonists & inhibitors , Periplasmic Proteins/antagonists & inhibitors , Protease Inhibitors/pharmacology , Allosteric Regulation , Antineoplastic Agents/chemistry , Escherichia coli , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/metabolism , High-Temperature Requirement A Serine Peptidase 1/chemistry , High-Temperature Requirement A Serine Peptidase 1/metabolism , Humans , Periplasmic Proteins/chemistry , Periplasmic Proteins/metabolism , Protease Inhibitors/chemistry , Protein Binding , Serine Endopeptidases/chemistry , Serine Endopeptidases/metabolism
6.
Biochem Biophys Res Commun ; 516(1): 157-162, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31202460

ABSTRACT

Chlamydia (C.) trachomatis, characterized by a unique biphasic life cycle, is an obligate intracellular bacterial pathogen which is responsible for the highest number of sexually transmitted bacterial infections globally. However, its pathogenic mechanisms have not been fully elucidated because of its unique developmental cycle and obligate intracellular nature. High temperature requirement (HtrA), a critical protease and chaperone, has been previously demonstrated to be essential for several functions and the replicative phase in the C. trachomatis developmental cycle. In the current study, we designed and synthesized a novel peptidomimetic inhibitor targeting C. trachomatis HtrA (CtHtrA) using homology modeling and chemical synthesis. The inhibitor was tested in chlamydia in the mid-replicative phase and resulted in a significant loss of viable infectious progeny and diminishing inclusion size and number at a relatively low concentration. This finding not only indicates that CtHtrA plays a critical role during the replicative phase of the chlamydial developmental cycle but also reveals a useful target for the design of novel anti-chlamydial agents.


Subject(s)
Anti-Bacterial Agents/pharmacology , Chlamydia Infections/drug therapy , Chlamydia trachomatis/drug effects , Peptidomimetics/pharmacology , Protease Inhibitors/pharmacology , Vacuoles/drug effects , Anti-Bacterial Agents/chemistry , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Chlamydia Infections/metabolism , Chlamydia trachomatis/chemistry , Chlamydia trachomatis/enzymology , Chlamydia trachomatis/growth & development , Drug Design , HeLa Cells , High-Temperature Requirement A Serine Peptidase 1/antagonists & inhibitors , High-Temperature Requirement A Serine Peptidase 1/chemistry , High-Temperature Requirement A Serine Peptidase 1/metabolism , Humans , Molecular Docking Simulation , Peptidomimetics/chemistry , Protease Inhibitors/chemistry , Vacuoles/metabolism
7.
Acta Crystallogr D Struct Biol ; 74(Pt 9): 906-921, 2018 Sep 01.
Article in English | MEDLINE | ID: mdl-30198900

ABSTRACT

High-temperature requirement A (HtrA) proteins, which are members of the heat-shock-induced serine protease family, are involved in extracytoplasmic protein quality control and bacterial survival strategies under stress conditions, and are associated with the virulence of several pathogens; they are therefore major drug targets. Mycobacterium tuberculosis possesses three putative HtrAs: HtrA1 (Rv1223), HtrA2 (Rv0983) and HtrA3 (Rv0125). Each has a cytoplasmic region, a transmembrane helix and a periplasmic region. Here, the crystal structure of the periplasmic region consisting of a protease domain (PD) and a PDZ domain from an M. tuberculosis HtrA1 mutant (mHtrA1S387A) is reported at 2.7 Šresolution. Although the mHtrA1S387A PD shows structural features similar to those of other HtrAs, its loops, particularly L3 and LA, display different conformations. Loop L3 communicates between the PDs of the trimer and the PDZ domains and undergoes a transition from an active to an inactive conformation, as reported for an equivalent HtrA (DegS). Loop LA, which is responsible for higher oligomer formation owing to its length (50 amino acids) in DegP, is very short in mHtrA1S387A (five amino acids), as in mHtrA2 (also five amino acids), and therefore lacks essential interactions for the formation of higher oligomers. Notably, a well ordered loop known as the insertion clamp in the PDZ domain interacts with the protease domain of the adjacent molecule, which possibly aids in the stabilization of a trimeric functional unit of this enzyme. The three-dimensional structure of mHtrA1S387A presented here will be useful in the design of enzyme-specific antituberculosis inhibitors.


Subject(s)
High-Temperature Requirement A Serine Peptidase 1/chemistry , High-Temperature Requirement A Serine Peptidase 1/metabolism , Mycobacterium tuberculosis/enzymology , PDZ Domains , Amino Acid Sequence , Models, Molecular , Protein Structure, Tertiary , Sequence Homology , Structure-Activity Relationship , Temperature
8.
J Mol Recognit ; 31(6): e2698, 2018 06.
Article in English | MEDLINE | ID: mdl-29266444

ABSTRACT

The human HtrA family of serine proteases (HtrA1, HtrA2, HtrA3, and HtrA4) are the key enzymes associated with pregnancy and closely related to the development and progression of many pathological events. Previously, it was found that halogen substitution at the indole moiety of peptide Trp-1 residue can form a geometrically satisfactory halogen bond with the Drosophila discs large, zona occludens-1 (PDZ) domain of HtrA proteases. Here, we attempt to systematically investigate the effect of substitution with 4 halogen types and 2 indole positions on the binding affinity and specificity of peptide ligands to the 4 HtrA PDZ domains. The complex structures, interaction energies, halogen-bonding strength, and binding affinity of domain-peptide systems were modeled, analyzed, and measured via computational modeling and fluorescence-based assay. It is revealed that there is a compromise between the local rearrangement of halogen bond involving different halogen atoms and the global optimization of domain-peptide interaction; the substitution position is fundamentally important for peptide-binding affinity, while the halogen type can effectively shift peptide selectivity between the 4 domains. The HtrA1-PDZ and HtrA4-PDZ as well as HtrA2-PDZ and HtrA3-PDZ respond similarly to different halogen substitutions of peptide; -Br substitution at R2-position and -I substitution at R4-position are most effective in improving peptide selectivity for HtrA1-PDZ/HtrA4-PDZ and HtrA2-PDZ/HtrA3-PDZ, respectively; -F substitution would not address substantial effect on peptide selectivity for all the 4 domains. Consequently, the binding affinities of a native peptide ligand DSRIWWV-COOH as well as its 4 R2-halogenated counterparts were determined as 1.9, 1.4, 0.5, 0.27, and 0.92 µM, which are basically consistent with computational analysis. This study would help to rationally design selective peptide inhibitors of HtrA family members by using different halogen substitutions.


Subject(s)
Peptides/genetics , Crystallography, X-Ray , Female , Halogens/chemistry , High-Temperature Requirement A Serine Peptidase 1/chemistry , High-Temperature Requirement A Serine Peptidase 1/genetics , High-Temperature Requirement A Serine Peptidase 2/chemistry , High-Temperature Requirement A Serine Peptidase 2/genetics , Humans , Indoles/chemistry , Ligands , PDZ Domains/genetics , Peptides/chemistry , Pregnancy , Protein Binding , Quantum Theory , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Serine Proteases/chemistry , Serine Proteases/genetics
9.
Sci Rep ; 7(1): 14804, 2017 11 01.
Article in English | MEDLINE | ID: mdl-29093542

ABSTRACT

The human protease family HtrA is responsible for preventing protein misfolding and mislocalization, and a key player in several cellular processes. Among these, HtrA1 is implicated in several cancers, cerebrovascular disease and age-related macular degeneration. Currently, HtrA1 activation is not fully characterized and relevant for drug-targeting this protease. Our work provides a mechanistic step-by-step description of HtrA1 activation and regulation. We report that the HtrA1 trimer is regulated by an allosteric mechanism by which monomers relay the activation signal to each other, in a PDZ-domain independent fashion. Notably, we show that inhibitor binding is precluded if HtrA1 monomers cannot communicate with each other. Our study establishes how HtrA1 trimerization plays a fundamental role in proteolytic activity. Moreover, it offers a structural explanation for HtrA1-defective pathologies as well as mechanistic insights into the degradation of complex extracellular fibrils such as tubulin, amyloid beta and tau that belong to the repertoire of HtrA1.


Subject(s)
High-Temperature Requirement A Serine Peptidase 1/chemistry , Protein Multimerization , Proteolysis , Allosteric Regulation , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , High-Temperature Requirement A Serine Peptidase 1/genetics , High-Temperature Requirement A Serine Peptidase 1/metabolism , Humans , Protein Domains , Structure-Activity Relationship , Tubulin/chemistry , Tubulin/genetics , Tubulin/metabolism , tau Proteins/chemistry , tau Proteins/genetics , tau Proteins/metabolism
11.
J Am Chem Soc ; 138(30): 9473-8, 2016 08 03.
Article in English | MEDLINE | ID: mdl-27379525

ABSTRACT

Apolipoprotein E (ApoE) belongs to a large class of proteins that solubilize lipids for physiological transport. Humans have three different APOE alleles, APOE ε2, APOE ε3, and APOE ε4, and genetic studies identified ApoE4 as the strongest genetic risk factor for Alzheimer's disease (AD). People who are homozygous for ApoE4 (i.e., ApoE4/E4) are an order of magnitude more likely to develop late-onset AD (LOAD) than ApoE3/E3 carriers. Several differences between ApoE3 and ApoE4 may contribute to AD including the observation that ApoE4 is degraded to a greater extent than ApoE3 in the human brain. Experiments with high-temperature requirement serine peptidase A1 (HtrA1), which is found in the nervous system, demonstrate that HtrA1 is an allele-selective ApoE-degrading enzyme that degrades ApoE4 more quickly than ApoE3. This activity is specific to HtrA1, as similar assays with HtrA2 showed minimal ApoE4 proteolysis and trypsin had no preference between ApoE4 and ApoE3. HtrA1 has also been reported to cleave the tau protein (Tau) and the amyloid protein precursor (APP) to hinder the formation of toxic amyloid deposits associated with AD. Competition assays with ApoE4, ApoE3, and Tau revealed that ApoE4 inhibits Tau degradation. Thus, the identification of ApoE4 as an in vitro HtrA1 substrate suggests a potential biochemical mechanism that links ApoE4 regulation of AD proteins such as Tau.


Subject(s)
Alleles , Apolipoproteins E/genetics , High-Temperature Requirement A Serine Peptidase 1/metabolism , Proteolysis , HEK293 Cells , High-Temperature Requirement A Serine Peptidase 1/chemistry , Humans , Models, Molecular , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL
...