Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Matrix Biol ; 129: 15-28, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38548090

RESUMO

Cathepsin K (CtsK) is a cysteine protease with potent collagenase activity. CtsK is highly expressed by bone-resorbing osteoclasts and plays an essential role in resorption of bone matrix. Although CtsK is known to bind heparan sulfate (HS), the structural details of the interaction, and how HS regulates the biological functions of CtsK, remains largely unknown. In this report, we discovered that HS is a multifaceted regulator of the structure and function of CtsK. Structurally, HS forms a highly stable complex with CtsK and induces its dimerization. Co-crystal structures of CtsK with bound HS oligosaccharides reveal the location of the HS binding site and suggest how HS may support dimerization. Functionally, HS plays a dual role in regulating the enzymatic activity of CtsK. While it preserves the peptidase activity of CtsK by stabilizing its active conformation, it inhibits the collagenase activity of CtsK in a sulfation level-dependent manner. These opposing effects can be explained by our finding that the HS binding site is remote from the active site, which allows HS to specifically inhibit the collagenase activity without affecting the peptidase activity. At last, we show that structurally defined HS oligosaccharides effectively block osteoclast resorption of bone in vitro without inhibiting osteoclast differentiation, which suggests that HS-based oligosaccharide might be explored as a new class of selective CtsK inhibitor for many diseases involving exaggerated bone resorption.


Assuntos
Catepsina K , Colagenases , Heparitina Sulfato , Osteoclastos , Catepsina K/metabolismo , Catepsina K/antagonistas & inibidores , Catepsina K/química , Catepsina K/genética , Heparitina Sulfato/metabolismo , Heparitina Sulfato/química , Colagenases/metabolismo , Humanos , Animais , Osteoclastos/metabolismo , Osteoclastos/efeitos dos fármacos , Sítios de Ligação , Camundongos , Cristalografia por Raios X , Reabsorção Óssea/metabolismo , Reabsorção Óssea/tratamento farmacológico , Ligação Proteica , Domínio Catalítico , Modelos Moleculares , Multimerização Proteica
2.
J Med Chem ; 64(18): 13793-13806, 2021 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-34473502

RESUMO

The cysteine protease cathepsin K is a target for the treatment of diseases associated with high bone turnover. Cathepsin K is mainly expressed in osteoclasts and responsible for the destruction of the proteinaceous components of the bone matrix. We designed various fluorescent activity-based probes (ABPs) and their precursors that bind to and inactivate cathepsin K. ABP 25 exhibited extraordinary potency (kinac/Ki = 35,300 M-1s-1) and selectivity for human cathepsin K. Crystal structures of cathepsin K in complex with ABP 25 and its nonfluorescent precursor 21 were determined to characterize the binding mode of this new type of acrylamide-based Michael acceptor with the particular orientation of the dibenzylamine moiety to the primed subsite region. The cyanine-5 containing probe 25 allowed for sensitive detection of cathepsin K, selective visualization in complex proteomes, and live cell imaging of a human osteosarcoma cell line, underlining its applicability in a pathophysiological environment.


Assuntos
Acrilamidas/química , Catepsina K/antagonistas & inibidores , Inibidores de Cisteína Proteinase/química , Corantes Fluorescentes/química , Acrilamidas/síntese química , Acrilamidas/metabolismo , Domínio Catalítico , Catepsina K/química , Catepsina K/metabolismo , Linhagem Celular Tumoral , Inibidores de Cisteína Proteinase/síntese química , Inibidores de Cisteína Proteinase/metabolismo , Desenho de Fármacos , Corantes Fluorescentes/síntese química , Corantes Fluorescentes/metabolismo , Humanos , Microscopia Confocal , Microscopia de Fluorescência , Ligação Proteica
3.
Bioorg Med Chem ; 28(15): 115597, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32631567

RESUMO

Cathepsin K (CatK) is a cysteine protease known for its potent collagenolytic activity, being recognized as an important target to the development of therapies for the treatment of bone disorders. Epoxypeptidomimetics have been reported as potent inhibitors of cathepsins, thus in this work we present a green synthesis of new peptidomimetics by using a one-pot asymmetric epoxidation/Ugi multicomponent reaction. The compounds were evaluated against CatK showing selectivity when compared with cathepsin L, with an inhibition profile in the low micromolar IC50 range. Investigation of the mechanism of action carried out for compounds LSPN428 and LSPN694 suggested a mixed inhibition mode and docking studies allowed a better understanding about interactions of inhibitors with the enzyme.


Assuntos
Catepsina K/antagonistas & inibidores , Inibidores de Cisteína Proteinase/química , Compostos de Epóxi/química , Peptidomiméticos/química , Domínio Catalítico , Catepsina K/química , Catepsina K/metabolismo , Inibidores de Cisteína Proteinase/síntese química , Inibidores de Cisteína Proteinase/metabolismo , Compostos de Epóxi/síntese química , Compostos de Epóxi/metabolismo , Química Verde , Humanos , Simulação de Acoplamento Molecular , Estrutura Molecular , Peptidomiméticos/síntese química , Peptidomiméticos/metabolismo , Ligação Proteica , Relação Estrutura-Atividade
4.
Fish Shellfish Immunol ; 98: 499-507, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32001355

RESUMO

Cathepsin K belongs to the family of cysteine cathepsins. It is well known that the cysteine cathepsins participate in various physiological processes and host immune defense in mammals. However, in teleost fish, the function of cathepsin K is very limited. In the present study, a cathepsin K homologue (SsCTSK) from the teleost black rockfish (Sebastes schlegelii) was identified and examined at expression and functional levels. In silico analysis showed that three domains, including signal peptide, cathepsin propeptide inhibitor I29 domain, and functional domain Pept_C1, are existed in SsCTSK. SsCTSK also possesses a peptidase domain with three catalytically essential residues (Cys25, His162 and Asn183). Phylogenetic profiling indicated that SsCTSK was evolutionally close to the cathepsin K of other teleost fish. Expression of SsCTSK occurred in multiple tissues and was induced by bacterial infection. Purified recombinant SsCTSK (rSsCTSK) exhibited apparent maximal peptidase activity at 45 °C, and its enzymatic activity was remarkably declined in the presence of the cathepsin inhibitor E-64. Moreover, rSsCTSK possesses the ability to bind with PAMPs and bacteria. Finally, knockdown of SsCTSK expression facilitated bacterial invasion in black rockfish. Collectively, these results indicated that SsCTSK functions as a cysteine protease and may serves as a target for pathogen manipulation of host defense system.


Assuntos
Catepsina K/química , Proteínas de Peixes/metabolismo , Regulação da Expressão Gênica/imunologia , Perciformes , Vibrioses/veterinária , Vibrio , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , Doenças dos Peixes/imunologia , Doenças dos Peixes/microbiologia , Proteínas de Peixes/química , Proteínas de Peixes/genética , Filogenia , Vibrioses/imunologia , Vibrioses/microbiologia
5.
Biochem J ; 477(1): 227-242, 2020 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-31860022

RESUMO

We have previously determined that the elastolytic activities of cathepsins (Cat) K and V require two exosites sharing the same structural localization on both enzymes. The structural features involved in the elastolytic activity of CatS have not yet been identified. We first mutated the analogous CatK and V putative exosites of CatS into the elastolytically inactive CatL counterparts. The modification of the exosite 1 did not affect the elastase activity of CatS whilst mutation of the Y118 of exosite 2 decreased the cleavage of elastin by ∼70% without affecting the degradation of other macromolecular substrates (gelatin, thyroglobulin). T06, an ectosteric inhibitor that disrupt the elastolytic activity of CatK, blocked ∼80% of the elastolytic activity of CatS without blocking the cleavage of gelatin and thyroglobulin. Docking studies showed that T06 preferentially interacts with a binding site located on the Right domain of the enzyme, outside of the active site. The structural examination of this binding site showed that the loop spanning the L174N175G176K177 residues of CatS is considerably different from that of CatL. Mutation of this loop into the CatL-like equivalent decreased elastin degradation by ∼70% and adding the Y118 mutation brought down the loss of elastolysis to ∼80%. In addition, the Y118 mutation selectively reduced the cleavage of the basement membrane component laminin by ∼50%. In summary, our data show that the degradation of elastin by CatS requires two exosites where one of them is distinct from those of CatK and V whilst the cleavage of laminin requires only one exosite.


Assuntos
Catepsinas/química , Elastina/metabolismo , Animais , Sítios de Ligação , Domínio Catalítico , Catepsina K/química , Cisteína Endopeptidases/química , Humanos , Camundongos Endogâmicos C57BL , Especificidade por Substrato
6.
Org Lett ; 21(2): 508-512, 2019 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-30628449

RESUMO

A total synthesis of N-desmethyl thalassospiramide C, a unique strained macrocyclic proteobacterial depsipeptide, enabled a detailed crystallographic study of its covalent complex with cathepsin K, a member of a medicinally important family of cysteine proteases. The study provides support for the mechanism of action, and the insight gained can be used for structure-based drug design targeting these calpain proteases.


Assuntos
Catepsina K/química , Inibidores de Cisteína Proteinase/síntese química , Cisteína/química , Serina Endopeptidases/química , Inibidores de Cisteína Proteinase/química , Estrutura Molecular
7.
PLoS One ; 14(1): e0211227, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30682119

RESUMO

Trypanosoma cruzi is the causative agent of Chagas disease, a neglected infection affecting millions of people in tropical regions. There are several chemotherapeutic agents for the treatment of this disease, but most of them are highly toxic and generate resistance. Currently, the development of allosteric inhibitors constitutes a promising research field, since it can improve the accessibility to more selective and less toxic medicines. To date, the allosteric drugs prediction is a state-of-the-art topic in rational structure-based computational design. In this work, a simulation strategy was developed for computational discovery of allosteric inhibitors, and it was applied to cruzain, a promising target and the major cysteine protease of T. cruzi. Molecular dynamics simulations, binding free energy calculations and network-based modelling of residue interactions were combined to characterize and compare molecular distinctive features of the apo form and the cruzain-allosteric inhibitor complexes. By using geometry-based criteria on trajectory snapshots, we predicted two main allosteric sites suitable for drug targeting. The results suggest dissimilar mechanisms exerted by the same allosteric site when binding different potential allosteric inhibitors. Finally, we identified the residues involved in suboptimal paths linking the identified site and the orthosteric site. The present study constitutes the first approximation to the design of cruzain allosteric inhibitors and may serve for future pharmacological intervention. Here, no major effects on active site structure were observed due to compound binding (modification of distance and angles between catalytic residues), which indicates that allosteric regulation in cruzain might be mediated via alterations of its dynamical properties similarly to allosteric inhibition of human cathepsin K (HCatK). The current findings are particularly relevant for the design of allosteric modulators of papain-like cysteine proteases.


Assuntos
Cisteína Endopeptidases/química , Cisteína Endopeptidases/metabolismo , Inibidores de Cisteína Proteinase/química , Proteínas de Protozoários/química , Proteínas de Protozoários/metabolismo , Trypanosoma cruzi/metabolismo , Regulação Alostérica/efeitos dos fármacos , Domínio Catalítico/efeitos dos fármacos , Catepsina K/química , Catepsina K/efeitos dos fármacos , Desenho Assistido por Computador , Inibidores de Cisteína Proteinase/farmacologia , Humanos , Modelos Moleculares , Simulação de Dinâmica Molecular , Conformação Proteica , Relação Estrutura-Atividade , Tripanossomicidas/química , Tripanossomicidas/farmacologia , Trypanosoma cruzi/efeitos dos fármacos
8.
Biochem J ; 476(3): 499-512, 2019 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-30622151

RESUMO

Cathepsin K (CatK) is a cysteine protease and drug target for skeletal disorders that is known for its potent collagenase and elastase activity. The formation of oligomeric complexes of CatK in the presence of glycosaminoglycans has been associated with its collagenase activity. Inhibitors that disrupt these complexes can selectively block the collagenase activity without interfering with the other regulatory proteolytic activities of the enzyme. Here, we have developed a fluorescence polarization (FP) assay to screen 4761 compounds for substrate-specific ectosteric collagenase inhibitors of CatK. A total of 38 compounds were identified that block the collagenase activity without interfering with the hydrolysis of active site substrates such as the synthetic peptide substrate, benzyloxycarbonyl-Phe-Arg-7-amido-4-methylcoumarin, and gelatin. The identified inhibitors can be divided into two main classes, negatively charged and polyaromatic compounds which suggest the binding to different ectosteric sites. Two of the inhibitors were highly effective in preventing the bone-resorption activity of CatK in osteoclasts. Interestingly, some of the ectosteric inhibitors were capable of differentiating between the collagenase and elastase activity of CatK depending on the ectosteric site utilized by the compound. Owing to their substrate-specific selectivity, ectosteric inhibitors represent a viable alternative to side effect-prone active site-directed inhibitors.


Assuntos
Catepsina K/antagonistas & inibidores , Peptídeos/química , Inibidores de Proteases/química , Animais , Catepsina K/química , Catepsina K/metabolismo , Bovinos , Humanos , Osteoclastos/enzimologia , Especificidade por Substrato
9.
FEBS J ; 285(22): 4265-4280, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30199612

RESUMO

Human cathepsin K (CTSK) is a collagenolytic lysosomal cysteine protease that plays an important role in bone turnover. Mutation in CTSK gene is associated with loss of collagenolytic activity of CTSK leading to an autosomal recessive bone disorder called pycnodysostosis. Although a number of pycnodysostotic missense mutations have been reported, underlying mechanism of the disease is not clear. In this study, we investigated in vitro six recombinant pycnodysostosis-related mutants of human CTSK (G79E, I249T, G243E, G303E, G319C and Q187P). While all the mutants, like wild-type, show similar high levels of expression in Escherichia coli, four of them (G79E, G303E, G319C and Q187P) are inactive, unstable and spontaneously degrade during purification process. In contrast, proteolytic/collagenolytic activity, zymogen activation kinetics and stability of G243E and I249T mutants are nominally affected. Crystal structure of I249T at 1.92 Å resolution shows that the mutation in R-domain causes conformational changes of a surface loop in the L-domain although the catalytic cleft remains unaltered. Molecular simulation, normal mode analysis and fluorescence lifetime measurement eliminated the possibility that the change in L-domain surface loop orientation is a crystallization artefact. CD-based thermal melting profile indicates that stability of I249T is significantly higher than wild-type. Our studies first time reports that pycnodysostosis-related mutations do not always lead to complete loss of general proteolytic activity or specific collagenolytic activity of CTSK. The first crystal structure of a pycnodysostotic mutant (I249T) provides critical information that may pave new avenues towards understanding the disease at molecular level. DATABASE: The atomic co-ordinates and structure factors for I249T mutant of human CTSK (codes 5Z5O) have been deposited in the Protein Data Bank (http://wwpdb.org/).


Assuntos
Catepsina K/química , Catepsina K/metabolismo , Mutação , Picnodisostose/genética , Sequência de Aminoácidos , Catálise , Catepsina K/genética , Cristalografia por Raios X , Análise Mutacional de DNA , Humanos , Modelos Moleculares , Conformação Proteica , Homologia de Sequência
10.
Biomed Mater ; 13(3): 035012, 2018 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-29442071

RESUMO

Bone tissue engineering with cell-scaffold constructs has been attracting a lot of attention, in particular as a tool for the efficient guiding of new tissue formation. However, the majority of the current strategies used to evaluate novel biomaterials focus on osteoblasts and bone formation, while osteoclasts are often overlooked. Consequently, there is limited knowledge on the interaction between osteoclasts and biomaterials. In this study, the ability of spongy-like gellan gum and hydroxyapatite-reinforced gellan gum hydrogels to support osteoclastogenesis was investigated in vitro. First, the spongy-like gellan gum and hydroxyapatite-reinforced gellan gum hydrogels were characterized in terms of microstructure, water uptake and mechanical properties. Then, bone marrow cells isolated from the long bones of mice and cultured in spongy-like hydrogels were treated with 1,25-dihydroxyvitamin D3 to promote osteoclastogenesis. It was shown that the addition of HAp to spongy-like gellan gum hydrogels enables the formation of larger pores and thicker walls, promoting an increase in stiffness. Hydroxyapatite-reinforced spongy-like gellan gum hydrogels support the formation of the aggregates of tartrate-resistant acid phosphatase-stained cells and the expression of genes encoding DC-STAMP and Cathepsin K, suggesting the differentiation of bone marrow cells into pre-osteoclasts. The hydroxyapatite-reinforced spongy-like gellan gum hydrogels developed in this work show promise for future use in bone tissue scaffolding applications.


Assuntos
Hidrogéis/química , Osteoclastos/citologia , Polissacarídeos Bacterianos/química , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Materiais Biocompatíveis/química , Células da Medula Óssea/citologia , Osso e Ossos/citologia , Calcitriol/química , Catepsina K/química , Diferenciação Celular , Sobrevivência Celular , Células Cultivadas , Durapatita/química , Masculino , Camundongos , Osteoblastos/citologia
11.
Cell Biochem Biophys ; 76(1-2): 219-229, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29322360

RESUMO

Pro-domain of a cysteine cathepsin contains a highly conserved Ex2Rx2Fx2Nx3Ix3N (ERFNIN) motif. The zymogen structure of cathepsins revealed that the Arg(R) residue of the motif is a central residue of a salt-bridge/H-bond network, stabilizing the scaffold of the pro-domain. Importance of the arginine is also demonstrated in studies where a single mutation (Arg → Trp) in human lysosomal cathepsin K (hCTSK) is linked to a bone-related genetic disorder "Pycnodysostosis". In the present study, we have characterized in vitro Arg → Trp mutant of hCTSK and the same mutant of hCTSL. The R → W mutant of hCTSK revealed that this mutation leads to an unstable zymogen that is spontaneously activated and auto-proteolytically degraded rapidly. In contrast, the same mutant of hCTSL is sufficiently stable and has proteolytic activity almost like its wild-type counterpart; however it shows an altered zymogen activation condition in terms of pH, temperature and time. Far and near UV circular dichroism and intrinsic tryptophan fluorescence experiments have revealed that the mutation has minimal effect on structure of the protease hCTSL. Molecular modeling studies shows that the mutated Trp31 in hCTSL forms an aromatic cluster with Tyr23 and Trp30 leading to a local stabilization of pro-domain and supplements the loss of salt-bridge interaction mediated by Arg31 in wild-type. In hCTSK-R31W mutant, due to presence of a non-aromatic Ser30 residue such interaction is not possible and may be responsible for local instability. These differences may cause detrimental effects of R31W mutation on the regulation of hCTSK auto-activation process compared to altered activation process in hCTSL.


Assuntos
Arginina/metabolismo , Catepsina K/metabolismo , Catepsina L/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Arginina/química , Catepsina K/química , Catepsina K/genética , Catepsina L/química , Catepsina L/genética , Dicroísmo Circular , Precursores Enzimáticos/química , Precursores Enzimáticos/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Cinética , Modelos Moleculares , Mutagênese Sítio-Dirigida , Estrutura Terciária de Proteína , Espectrometria de Fluorescência , Especificidade por Substrato
12.
Protein Sci ; 27(3): 714-724, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29266558

RESUMO

Fibrin clot formation is a proteolytic cascade of events with thrombin and plasmin identified as the main proteases cleaving fibrinogen precursor, and the fibrin polymer, respectively. Other proteases may be involved directly in fibrin(ogen) cleavage, clot formation, and resolution, or in the degradation of fibrin-based scaffolds emerging as useful tools for tissue engineered constructs. Here, cysteine cathepsins are investigated for their putative ability to hydrolyze fibrinogen, since they are potent proteases, first identified in lysosomal protein degradation and known to participate in extracellular proteolysis. To further explore this, we used two independent computational technqiues, molecular docking and bioinformatics sequence analysis (PACMANS), to predict potential binding interactions and sites of hydrolysis between cathepsins K, L, and S and fibrinogen. By comparing the results from these two objective, computational methods, it was determined that cathepsins K, L, and S do bind and cleave fibrinogen α, ß, and γ chains at similar and unique sites. These differences were visualized experimentally by the unique cleaved fibrinogen banding patterns after incubation with each of the cathepsins, separately. In conclusion, human cysteine cathepsins K, L, and S are a new class of proteases that should be considered during fibrin(ogen) degradation studies both for disease processes where coagulation is a concern, and also in the implementation and design of bioengineered systems.


Assuntos
Catepsinas/metabolismo , Biologia Computacional/métodos , Fibrinogênio/química , Fibrinogênio/metabolismo , Sítios de Ligação , Catepsina K/química , Catepsina K/metabolismo , Catepsina L/química , Catepsina L/metabolismo , Catepsinas/química , Humanos , Modelos Moleculares , Simulação de Acoplamento Molecular , Ligação Proteica , Conformação Proteica , Proteólise
13.
PLoS One ; 12(10): e0186869, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29088253

RESUMO

Cathepsin K (CatK) is a cysteine protease that plays an important role in mammalian intra- and extracellular protein turnover and is known for its unique and potent collagenase activity. Through studies on the mechanism of its collagenase activity, selective ectosteric sites were identified that are remote from the active site. Inhibitors targeting these ectosteric sites are collagenase selective and do not interfere with other proteolytic activities of the enzyme. Potential ectosteric inhibitors were identified using a computational approach to screen the druggable subset of and the entire 281,987 compounds comprising Chemical Repository library of the National Cancer Institute-Developmental Therapeutics Program (NCI-DTP). Compounds were scored based on their affinity for the ectosteric site. Here we compared the scores of three individual molecular docking methods with that of a composite score of all three methods together. The composite docking method was up to five-fold more effective at identifying potent collagenase inhibitors (IC50 < 20 µM) than the individual methods. Of 160 top compounds tested in enzymatic assays, 28 compounds revealed blocking of the collagenase activity of CatK at 100 µM. Two compounds exhibited IC50 values below 5 µM corresponding to a molar protease:inhibitor concentration of <1:12. Both compounds were subsequently tested in osteoclast bone resorption assays where the most potent inhibitor, 10-[2-[bis(2-hydroxyethyl)amino]ethyl]-7,8-diethylbenzo[g]pteridine-2,4-dione, (NSC-374902), displayed an inhibition of bone resorption with an IC50-value of approximately 300 nM and no cell toxicity effects.


Assuntos
Catepsina K/antagonistas & inibidores , Inibidores de Cisteína Proteinase/farmacologia , Simulação de Acoplamento Molecular/métodos , Regulação Alostérica , Sítio Alostérico , Sítios de Ligação , Domínio Catalítico , Catepsina K/química , Catepsina K/metabolismo , Células Cultivadas , Colagenases/química , Colagenases/metabolismo , Inibidores de Cisteína Proteinase/química , Inibidores de Cisteína Proteinase/metabolismo , Humanos , Estrutura Molecular , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Ligação Proteica , Domínios Proteicos
14.
Sci Rep ; 7(1): 11459, 2017 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-28904354

RESUMO

Cathepsin X is a cysteine peptidase involved in the progression of cancer and neurodegenerative diseases. Targeting this enzyme with selective inhibitors opens a new possibility for intervention in several therapeutic areas. In this study triazole-based reversible and selective inhibitors of cathepsin X have been identified. Their selectivity and binding is enhanced when the 2,3-dihydrobenzo[b][1,4]dioxine moiety is present as the R1 substituent. Of a series of selected triazole-benzodioxine derivatives, compound 22 is the most potent inhibitor of cathepsin X carboxypeptidase activity (Ki = 2.45 ± 0.05 µM) with at least 100-fold greater selectivity in comparison to cathepsin B or other related cysteine peptidases. Compound 22 is not cytotoxic to prostate cancer cells PC-3 or pheochromocytoma PC-12 cells at concentrations up to 10 µM. It significantly inhibits the migration of tumor cells and increases the outgrowth of neurites, both processes being under the control of cathepsin X carboxypeptidase activity. Compound 22 and other characterized triazole-based inhibitors thus possess a great potential for further development resulting in several in vivo applications.


Assuntos
Catepsina K/antagonistas & inibidores , Inibidores de Cisteína Proteinase/farmacologia , Descoberta de Drogas , Animais , Catepsina K/química , Inibidores de Cisteína Proteinase/química , Descoberta de Drogas/métodos , Cinética , Espectroscopia de Ressonância Magnética , Estrutura Molecular , Crescimento Neuronal/efeitos dos fármacos , Células PC12 , Ligação Proteica , Ratos , Bibliotecas de Moléculas Pequenas , Relação Estrutura-Atividade
15.
PLoS One ; 12(8): e0182387, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28771551

RESUMO

Allosteric targeting is progressively gaining ground as a strategy in drug design. Its success, however, depends on our knowledge of the investigated system. In the case of the papain-like cysteine peptidase cathepsin K, a major obstacle in our understanding of allostery is represented by the lack of observable conformational change at the active site. This makes it difficult to understand how binding of effectors at known allosteric sites translates into modified enzyme activity. Herein, we address this issue by a computational approach based on experimental data. We analyze the conformational space of the papain-like family and the positioning of cathepsin K within it using principal component analysis and molecular dynamics simulations. We show that human cathepsin L-like endopeptidases (cathepsins L, K, S and V) adopt similar conformations which are distinct from their non-animal counterparts and other related peptidases. Molecular dynamics simulations show that the conformation of cathepsin K is influenced by known allosteric effectors, chondroitin sulfate and the small molecules NSC13345 and NSC94914. Importantly, all effectors affect the geometry of the active site around sites S1 and S2 that represent the narrowest part of the active site cleft and the major specificity determinant in papain-like endopeptidases. The effectors act by stabilizing pre-existing conformational states according to a two-state model and thereby facilitate or hinder the binding of substrate into the active site, as shown by molecular docking simulations. Comparison with other related enzymes shows that similar conformational variability and, by implication, allostery also exist in other papain-like endopeptidases.


Assuntos
Catepsina K/metabolismo , Regulação Alostérica , Sequência de Aminoácidos , Benzoatos/química , Benzoatos/metabolismo , Sítios de Ligação , Domínio Catalítico , Catepsina K/química , Sulfatos de Condroitina/química , Sulfatos de Condroitina/metabolismo , Humanos , Isoenzimas/química , Isoenzimas/metabolismo , Simulação de Dinâmica Molecular , Papaína/química , Papaína/metabolismo , Análise de Componente Principal , Alinhamento de Sequência , Especificidade por Substrato
16.
FEBS Lett ; 590(24): 4507-4518, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27859061

RESUMO

The cysteine peptidase cathepsin K is a potent collagenolytic enzyme and a promising target for the treatment of osteoporosis. Here, we characterize its allosteric fine-tuning via a recently identified allosteric site. We show that compound NSC94914 binds this site and acts as a specific partial inhibitor of the collagenolytic activity of cathepsin K. We link the functional differences between NSC94914 and known effectors (compound NSC11345 and glycosaminoglycans) to their different modes of interaction with the site. We characterize the allosteric site by site-directed mutagenesis and show that it is involved in specific regulation of the collagenolytic activity of cathepsin K.


Assuntos
Benzoatos/química , Catepsina K/química , Glicosaminoglicanos/química , Ácido Metilmalônico/análogos & derivados , Inibidores de Proteases/química , Regulação Alostérica , Sítio Alostérico , Catepsina K/antagonistas & inibidores , Catepsina K/genética , Catepsina K/metabolismo , Colágeno/química , Colágeno/metabolismo , Cristalografia por Raios X , Elastina/química , Elastina/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Humanos , Hidrólise , Cinética , Modelos Moleculares , Mutagênese Sítio-Dirigida , Mutação , Domínios Proteicos , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Relação Estrutura-Atividade , Especificidade por Substrato
17.
J Biol Chem ; 291(37): 19220-34, 2016 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-27422822

RESUMO

Kunitz-type (KT) protease inhibitors are low molecular weight proteins classically defined as serine protease inhibitors. We identified a novel secreted KT inhibitor associated with the gut and parenchymal tissues of the infective juvenile stage of Fasciola hepatica, a helminth parasite of medical and veterinary importance. Unexpectedly, recombinant KT inhibitor (rFhKT1) exhibited no inhibitory activity toward serine proteases but was a potent inhibitor of the major secreted cathepsin L cysteine proteases of F. hepatica, FhCL1 and FhCL2, and of human cathepsins L and K (Ki = 0.4-27 nm). FhKT1 prevented the auto-catalytic activation of FhCL1 and FhCL2 and formed stable complexes with the mature enzymes. Pulldown experiments from adult parasite culture medium showed that rFhKT1 interacts specifically with native secreted FhCL1, FhCL2, and FhCL5. Substitution of the unusual P1 Leu(15) within the exposed reactive loop of FhKT1 for the more commonly found Arg (FhKT1Leu(15)/Arg(15)) had modest adverse effects on the cysteine protease inhibition but conferred potent activity against the serine protease trypsin (Ki = 1.5 nm). Computational docking and sequence analysis provided hypotheses for the exclusive binding of FhKT1 to cysteine proteases, the importance of the Leu(15) in anchoring the inhibitor into the S2 active site pocket, and the inhibitor's selectivity toward FhCL1, FhCL2, and human cathepsins L and K. FhKT1 represents a novel evolutionary adaptation of KT protease inhibitors by F. hepatica, with its prime purpose likely in the regulation of the major parasite-secreted proteases and/or cathepsin L-like proteases of its host.


Assuntos
Catepsina K/antagonistas & inibidores , Catepsina L/antagonistas & inibidores , Inibidores de Cisteína Proteinase/química , Fasciola hepatica/química , Proteínas de Helminto/química , Animais , Catepsina K/química , Catepsina L/química , Humanos , Proteínas Recombinantes/química , Tripsina/química , Inibidores da Tripsina/química
18.
Ann N Y Acad Sci ; 1364: 52-61, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25800988

RESUMO

Mitochondrial dysfunction has emerged as an important factor in wide ranging human pathologies. We have previously defined a retrograde signaling pathway that originates from dysfunctional mitochondria (Mt-RS) and causes a global nuclear transcriptional reprograming as its end point. Mitochondrial dysfunction causing disruption of mitochondrial membrane potential and consequent increase in cytosolic calcium [Ca(2) ](c) activates calcineurin and the transcription factors NF-κB, NFAT, CREB, and C/EBPδ. In macrophages, this signaling complements receptor activator of nuclear factor kappa-B ligand (RANKL)-induced osteoclastic differentiation. Here, we show that the Mt-RS activated transcriptional coactivator heterogeneous ribonucleoprotein A2 (hnRNP A2) is induced by hypoxia in murine macrophages. We demonstrate that the cathepsin K gene (Ctsk), one of the key genes upregulated during osteoclast differentiation, is transcriptionally activated by Mt-RS factors. HnRNP A2 acts as a coactivator with nuclear transcription factors, cRel, and C/EBPδ for Ctsk promoter activation under hypoxic conditions. Notably, our study shows that hypoxia-induced activation of the stress target factors mediates effects similar to that of RANKL with regard to Ctsk activation. We therefore suggest that mitochondrial dysfunction and activation of Mt-RS, induced by various pathophysiologic conditions, is a potential risk factor for osteoclastogenesis and bone loss.


Assuntos
Catepsina K/metabolismo , Mitocôndrias/metabolismo , Osteoclastos/metabolismo , Osteogênese , Regiões Promotoras Genéticas , Transdução de Sinais , Animais , Proteína delta de Ligação ao Facilitador CCAAT/antagonistas & inibidores , Proteína delta de Ligação ao Facilitador CCAAT/genética , Proteína delta de Ligação ao Facilitador CCAAT/metabolismo , Catepsina K/antagonistas & inibidores , Catepsina K/química , Catepsina K/genética , Hipóxia Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/antagonistas & inibidores , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Genes Reporter , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/antagonistas & inibidores , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/genética , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/metabolismo , Camundongos , Mitocôndrias/enzimologia , Fatores de Transcrição NFATC/antagonistas & inibidores , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/metabolismo , Osteoclastos/citologia , Osteoclastos/enzimologia , Ligante RANK/metabolismo , Células RAW 264.7 , Interferência de RNA , RNA Mensageiro/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Ativação Transcricional
19.
BMC Biotechnol ; 15: 112, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26666739

RESUMO

BACKGROUND: Triple helical collagens are the most abundant structural protein in vertebrates and are widely used as biomaterials for a variety of applications including drug delivery and cellular and tissue engineering. In these applications, the mechanics of this hierarchically structured protein play a key role, as does its chemical composition. To facilitate investigation into how gene mutations of collagen lead to disease as well as the rational development of tunable mechanical and chemical properties of this full-length protein, production of recombinant expressed protein is required. RESULTS: Here, we present a human type II procollagen expression system that produces full-length procollagen utilizing a previously characterized human fibrosarcoma cell line for production. The system exploits a non-covalently linked fluorescence readout for gene expression to facilitate screening of cell lines. Biochemical and biophysical characterization of the secreted, purified protein are used to demonstrate the proper formation and function of the protein. Assays to demonstrate fidelity include proteolytic digestion, mass spectrometric sequence and posttranslational composition analysis, circular dichroism spectroscopy, single-molecule stretching with optical tweezers, atomic-force microscopy imaging of fibril assembly, and transmission electron microscopy imaging of self-assembled fibrils. CONCLUSIONS: Using a mammalian expression system, we produced full-length recombinant human type II procollagen. The integrity of the collagen preparation was verified by various structural and degradation assays. This system provides a platform from which to explore new directions in collagen manipulation.


Assuntos
Colágeno Tipo II/biossíntese , Colágeno Tipo II/genética , Eucariotos/genética , Eucariotos/metabolismo , Catepsina K/química , Catepsina K/metabolismo , Linhagem Celular Tumoral , Dicroísmo Circular , Células Clonais , Matriz Extracelular/metabolismo , Fibrossarcoma/genética , Fibrossarcoma/metabolismo , Fibrossarcoma/patologia , Humanos , Microscopia de Força Atômica , Pinças Ópticas , Pró-Colágeno/biossíntese , Pró-Colágeno/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfecção
20.
Proteomics ; 15(14): 2479-90, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25626674

RESUMO

Proteases are important effectors of numerous physiological and pathological processes. Reliable determination of a protease's specificity is crucial to understand protease function and to develop activity-based probes and inhibitors. During the last decade, various proteomic approaches for profiling protease substrate specificities were reported. Although most of these approaches can identify up to thousands of substrate cleavage events in a single experiment, they are often time consuming and methodologically challenging as some of these approaches require rather complex sample preparation procedures. For such reasons their application is often limited to those labs that initially introduced them. Here, we report on a fast and simple approach for proteomic profiling of protease specificities (fast profiling of protease specificity (FPPS)), which can be applied to complex protein mixtures. FPPS is based on trideutero-acetylation of novel N-termini generated by the action of proteases and subsequent peptide fractionation on Stage Tips containing ion-exchange and reverse phase chromatographic resins. FPPS can be performed in 2 days and does not require extensive fractionation steps. Using this approach, we have determined the specificity profiles of the cysteine cathepsins K, L and S. We further validated our method by comparing the results with the specificity profiles obtained by the N-terminal combined fractional diagonal chromatography method. This comparison pointed to almost identical substrate specificities for all three cathepsins and confirmed the reliability of the FPPS approach. All MS data have been deposited in the ProteomeXchange with identifiers PXD001536 and PXD001553 (http://proteomecentral.proteomexchange.org/dataset/PXD001536; http://proteomecentral.proteomexchange.org/dataset/PXD001553).


Assuntos
Catepsina K/metabolismo , Catepsina L/metabolismo , Catepsinas/metabolismo , Sequência de Aminoácidos , Catepsina K/química , Catepsina L/química , Catepsinas/química , Linhagem Celular Tumoral , Cromatografia Líquida/métodos , Humanos , Peptídeos/química , Peptídeos/metabolismo , Proteômica/métodos , Especificidade por Substrato , Espectrometria de Massas em Tandem/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA