Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros











Intervalo de año de publicación
1.
Int J Mol Sci ; 25(4)2024 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-38397107

RESUMEN

Predicting the potency of inhibitors is key to in silico screening of promising synthetic or natural compounds. Here we describe a predictive workflow that provides calculated inhibitory values, which concord well with empirical data. Calculations of the free interaction energy ΔG with the YASARA plugin FoldX were used to derive inhibition constants Ki from PDB coordinates of protease-inhibitor complexes. At the same time, corresponding KD values were obtained from the PRODIGY server. These results correlated well with the experimental values, particularly for serine proteases. In addition, analyses were performed for inhibitory complexes of cysteine and aspartic proteases, as well as of metalloproteases, whereby the PRODIGY data appeared to be more consistent. Based on our analyses, we calculated theoretical Ki values for trypsin with sunflower trypsin inhibitor (SFTI-1) variants, which yielded the more rigid Pro14 variant, with probably higher potency than the wild-type inhibitor. Moreover, a hirudin variant with an Arg1 and Trp3 is a promising basis for novel thrombin inhibitors with high potency. Further examples from antibody interaction and a cancer-related effector-receptor system demonstrate that our approach is applicable to protein interaction studies beyond the protease field.


Asunto(s)
Helianthus , Serina Endopeptidasas , Inhibidores de Tripsina/farmacología , Tripsina/metabolismo , Helianthus/metabolismo , Péptido Hidrolasas , Inhibidores de Proteasas/farmacología
2.
Int J Mol Sci ; 22(20)2021 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-34681855

RESUMEN

Site-specific incorporation of non-canonical amino acids (ncAAs) into proteins has emerged as a universal tool for systems bioengineering at the interface of chemistry, biology, and technology. The diversification of the repertoire of the genetic code has been achieved for amino acids with long and/or bulky side chains equipped with various bioorthogonal tags and useful spectral probes. Although ncAAs with relatively small side chains and similar properties are of great interest to biophysics, cell biology, and biomaterial science, they can rarely be incorporated into proteins. To address this gap, we report the engineering of PylRS variants capable of incorporating an entire library of aliphatic "small-tag" ncAAs. In particular, we performed mutational studies of a specific PylRS, designed to incorporate the shortest non-bulky ncAA (S-allyl-l-cysteine) possible to date and based on this knowledge incorporated aliphatic ncAA derivatives. In this way, we have not only increased the number of translationally active "small-tag" ncAAs, but also determined key residues responsible for maintaining orthogonality, while engineering the PylRS for these interesting substrates. Based on the known plasticity of PylRS toward different substrates, our approach further expands the reassignment capacities of this enzyme toward aliphatic amino acids with smaller side chains endowed with valuable functionalities.


Asunto(s)
Aminoacil-ARNt Sintetasas/metabolismo , Código Genético , Lisina/análogos & derivados , Methanosarcina barkeri/enzimología , Ingeniería de Proteínas , Aminoacil-ARNt Sintetasas/genética , Proteínas Arqueales/metabolismo , Lisina/metabolismo , Especificidad por Sustrato
3.
Int J Mol Sci ; 22(6)2021 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-33810118

RESUMEN

Experimental evidence for enzymatic mechanisms is often scarce, and in many cases inadvertently biased by the employed methods. Thus, apparently contradictory model mechanisms can result in decade long discussions about the correct interpretation of data and the true theory behind it. However, often such opposing views turn out to be special cases of a more comprehensive and superior concept. Molecular dynamics (MD) and the more advanced molecular mechanical and quantum mechanical approach (QM/MM) provide a relatively consistent framework to treat enzymatic mechanisms, in particular, the activity of proteolytic enzymes. In line with this, computational chemistry based on experimental structures came up with studies on all major protease classes in recent years; examples of aspartic, metallo-, cysteine, serine, and threonine protease mechanisms are well founded on corresponding standards. In addition, experimental evidence from enzyme kinetics, structural research, and various other methods supports the described calculated mechanisms. One step beyond is the application of this information to the design of new and powerful inhibitors of disease-related enzymes, such as the HIV protease. In this overview, a few examples demonstrate the high potential of the QM/MM approach for sophisticated pharmaceutical compound design and supporting functions in the analysis of biomolecular structures.


Asunto(s)
Simulación de Dinámica Molecular , Péptido Hidrolasas/química , Inhibidores de Proteasas/química , Teoría Cuántica , Algoritmos , Proteasas de Cisteína/química , Proteasas de Cisteína/metabolismo , Metaloproteasas/metabolismo , Estructura Molecular , Péptido Hidrolasas/metabolismo , Inhibidores de Proteasas/farmacología , Conformación Proteica , Serina Proteasas/química , Serina Proteasas/metabolismo , Termodinámica
4.
Cancer Metastasis Rev ; 38(3): 417-430, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31482487

RESUMEN

In the past decade, immune-based therapies such as monoclonal antibodies against tumor epitopes or immune checkpoint inhibitors have become an integral part of contemporary cancer treatment in many entities. However, a fundamental prerequisite for the success of such therapies is a sufficient trafficking of tumor-infiltrating lymphocytes into the tumor microenvironment. This infiltration is facilitated by chemokines, a group of about 50 small proteins capable of chemotactically guiding leukocytes. Proteolytic inactivation of chemokines leading to an impaired infiltration of immune effector cells appears to be an efficient immune escape mechanism of solid cancers.The CXCR3 and CX3CR1 chemokine receptor ligands CXCL9-11 and CX3CL1, respectively, are mainly responsible for the tumor-suppressive lymphocytic infiltration into the tumor micromilieu. Their structure explains the biochemical basis of their proteolytic cleavage, while in vivo data from mouse models and patient samples shed light on the corresponding processes in cancer. The emerging roles of proteases, e.g., matrix metalloproteinases, cathepsins, and dipeptidyl peptidase 4, in chemokine inactivation define new resistance mechanisms against immunotherapies and identify attractive new targets to enhance immune intervention in cancer.


Asunto(s)
Quimiocinas/inmunología , Quimiocinas/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Neoplasias/inmunología , Neoplasias/metabolismo , Secuencia de Aminoácidos , Animales , Receptor 1 de Quimiocinas CX3C/inmunología , Receptor 1 de Quimiocinas CX3C/metabolismo , Quimiocina CXCL10/química , Quimiocina CXCL10/inmunología , Quimiocina CXCL10/metabolismo , Quimiocina CXCL11/química , Quimiocina CXCL11/inmunología , Quimiocina CXCL11/metabolismo , Quimiocina CXCL9/química , Quimiocina CXCL9/inmunología , Quimiocina CXCL9/metabolismo , Quimiocinas/química , Humanos , Linfocitos Infiltrantes de Tumor/enzimología , Linfocitos Infiltrantes de Tumor/patología , Modelos Moleculares , Neoplasias/enzimología , Neoplasias/patología , Péptido Hidrolasas/metabolismo , Proteolisis , Receptores CXCR3/inmunología , Receptores CXCR3/metabolismo
5.
Biochimie ; 166: 52-76, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31505212

RESUMEN

Trypsin and chymotrypsin-like serine proteases from family S1 (clan PA) constitute the largest protease group in humans and more generally in vertebrates. The prototypes chymotrypsin, trypsin and elastase represent simple digestive proteases in the gut, where they cleave nearly any protein. Multidomain trypsin-like proteases are key players in the tightly controlled blood coagulation and complement systems, as well as related proteases that are secreted from diverse immune cells. Some serine proteases are expressed in nearly all tissues and fluids of the human body, such as the human kallikreins and kallikrein-related peptidases with specialization for often unique substrates and accurate timing of activity. HtrA and membrane-anchored serine proteases fulfill important physiological tasks with emerging roles in cancer. The high diversity of all family members, which share the tandem ß-barrel architecture of the chymotrypsin-fold in the catalytic domain, is conferred by the large differences of eight surface loops, surrounding the active site. The length of these loops alters with insertions and deletions, resulting in remarkably different three-dimensional arrangements. In addition, metal binding sites for Na+, Ca2+ and Zn2+ serve as regulatory elements, as do N-glycosylation sites. Depending on the individual tasks of the protease, the surface loops determine substrate specificity, control the turnover and allow regulation of activation, activity and degradation by other proteins, which are often serine proteases themselves. Most intriguingly, in some serine proteases, the surface loops interact as allosteric network, partially tuned by protein co-factors. Knowledge of these subtle and complicated molecular motions may allow nowadays for new and specific pharmaceutical or medical approaches.


Asunto(s)
Serina Endopeptidasas/química , Serina Endopeptidasas/fisiología , Animales , Sitios de Unión , Dominio Catalítico , Humanos , Modelos Moleculares , Conformación Proteica en Lámina beta , Relación Estructura-Actividad , Especificidad por Sustrato
6.
Expert Opin Ther Targets ; 22(9): 745-763, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30114962

RESUMEN

INTRODUCTION: Aberrant levels of kallikrein-related peptidases (KLK1-15) have been linked to cancer cell proliferation, invasion and metastasis. In ovarian cancer, the KLK proteolytic network has a crucial role in the tissue and tumor microenvironment. Publically available ovarian cancer genome and expression data from multiple patient cohorts show an upregulation of most KLKs. Areas covered: Here, we review the expression levels of all 15 members of this family in normal and ovarian cancer tissues, categorizing them into highly and moderately or weakly expressed KLKs, and their association with patient prognosis and survival. We summarize their tumor-biological functions determined in cell-based assays and xenograft models, further highlighting their suitability as cancer biomarkers and attractive candidates for drug development. Finally, we discuss some different pharmaceutical approaches, including peptide-based and small molecule inhibitors, cyclic peptides, depsipeptides, engineered natural inhibitors, antibodies, RNA/DNA-based aptamers, prodrugs, miRNA and siRNA. Expert opinion: In light of the results from clinical and tumor-biological studies, together with the available pharmaceutical tools, we suggest KLK4, KLK5, KLK6 and possibly KLK7 as preferred targets for inhibition in ovarian cancer.


Asunto(s)
Antineoplásicos/farmacología , Calicreínas/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Animales , Biomarcadores de Tumor/metabolismo , Desarrollo de Medicamentos/métodos , Femenino , Humanos , Terapia Molecular Dirigida , Neoplasias Ováricas/patología , Pronóstico , Microambiente Tumoral
7.
Biol Chem ; 399(9): 1009-1022, 2018 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-29975661

RESUMEN

Human kallikrein-related peptidases 3, 4, 11, and KLK2, the activator of KLK3/PSA, belong to the prostatic group of the KLKs, whose major physiological function is semen liquefaction during the fertilization process. Notably, these KLKs are upregulated in prostate cancer and are used as clinical biomarkers or have been proposed as therapeutic targets. However, this potential awaits a detailed characterization of these proteases. In order to study glycosylated prostatic KLKs resembling the natural proteases, we used Leishmania (LEXSY) and HEK293 cells for secretory expression. Both systems allowed the subsequent purification of soluble pro-KLK zymogens with correct propeptides and of the mature forms. Periodic acid-Schiff reaction, enzymatic deglycosylation assays, and mass spectrometry confirmed the glycosylation of these KLKs. Activation of glycosylated pro-KLKs 4 and 11 turned out to be most efficient by glycosylated KLK2 and KLK4, respectively. By comparing the glycosylated prostatic KLKs with their non-glycosylated counterparts from Escherichia coli, it was observed that the N-glycans stabilize the KLK proteases and change their activation profiles and their enzymatic activity to some extent. The functional role of glycosylation in prostate-specific KLKs could pave the way to a deeper understanding of their biology and to medical applications.


Asunto(s)
Calicreínas/metabolismo , Antígeno Prostático Específico/metabolismo , Serina Endopeptidasas/metabolismo , Clonación Molecular , Glicosilación , Células HEK293 , Humanos , Calicreínas/genética , Antígeno Prostático Específico/genética , Serina Endopeptidasas/genética
8.
Biol Chem ; 399(9): 997-1007, 2018 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-29883318

RESUMEN

In humans, three different trypsin-isoenzymes have been described. Of these, trypsin-3 appears to be functionally different from the others. In order to systematically study the specificity of the trypsin-isoenzymes, we utilized proteome-derived peptide libraries and quantitative proteomics. We found similar specificity profiles dominated by the well-characterized preference for cleavage after lysine and arginine. Especially, trypsin-1 slightly favored lysine over arginine in this position, while trypsin-3 did not discriminate between them. In the P1' position, which is the residue C-terminal to the cleavage site, we noticed a subtle enrichment of alanine and glycine for all three trypsins and for trypsin-3 there were additional minor P1' and P2' preferences for threonine and aspartic acid, respectively. These findings were confirmed by FRET peptide substrates showing different susceptibility to cleavage by different trypsins. The preference of trypsin-3 for aspartic acid in P2' is explained by salt bridge formation with the unique Arg193. This salt bridge enables and stabilizes a canonical oxyanion conformation by the amides of Ser195 and Arg193, thus manifesting a selective substrate-assisted catalysis. As trypsin-3 has been proposed to be a therapeutic target and marker for cancers, our results may aid the development of specific inhibitors for cancer therapy and diagnostic probes.


Asunto(s)
Tripsina/química , Tripsina/metabolismo , Secuencia de Aminoácidos , Colorantes Fluorescentes/química , Humanos , Isoenzimas/química , Isoenzimas/metabolismo , Modelos Moleculares , Alineación de Secuencia , Especificidad por Sustrato
9.
Matrix Biol ; 70: 84-101, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29572155

RESUMEN

Age-related macular degeneration (AMD) is the leading cause of irreversible blindness in the elderly population. Variants in the HTRA1-ARMS2 locus have been linked to increased AMD risk. In the present study we investigated the impact of elevated HtrA1 levels on the retina pigment epithelial (RPE) secretome using a polarized culture system. Upregulation of HtrA1 alters the abundance of key proteins involved in angiogenesis and extracellular matrix remodeling. Thrombospondin-1, an angiogenesis modulator, was identified as a substrate for HtrA1 using terminal amine isotope labeling of substrates in conjunction with HtrA1 specificity profiling. HtrA1 cleavage of thrombospondin-1 was further corroborated by in vitro cleavage assays and targeted proteomics together with small molecule inhibition of HtrA1. While thrombospondin-1 is anti-angiogenic, the proteolytically released N-terminal fragment promotes the formation of tube-like structure by endothelial cells. Taken together, our findings suggest a mechanism by which increased levels of HtrA1 may contribute to AMD pathogenesis. The proteomic data has been deposited to the ProteomeXchange Consortium via the PRIDE partner repository with the data set identifier. For quantitative secretome analysis, project accession: PXD007691, username: reviewer45093@ebi.ac.uk, password: 1FUpS6Yq. For TAILS analysis, project accession: PXD007139, username: reviewer76731@ebi.ac.uk, password: sNbMp7xK.


Asunto(s)
Inductores de la Angiogénesis/química , Serina Peptidasa A1 que Requiere Temperaturas Altas/metabolismo , Degeneración Macular/metabolismo , Fragmentos de Péptidos/química , Pigmentos Retinianos/metabolismo , Trombospondina 1/química , Anciano , Secuencia de Aminoácidos , Inductores de la Angiogénesis/aislamiento & purificación , Inductores de la Angiogénesis/farmacología , Medios de Cultivo Condicionados/química , Cámaras de Difusión de Cultivos , Impedancia Eléctrica , Células Epiteliales/metabolismo , Células Epiteliales/patología , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Serina Peptidasa A1 que Requiere Temperaturas Altas/genética , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Degeneración Macular/genética , Degeneración Macular/patología , Modelos Moleculares , Fragmentos de Péptidos/aislamiento & purificación , Fragmentos de Péptidos/farmacología , Cultivo Primario de Células , Proteolisis , Proteoma/genética , Proteoma/metabolismo , Pigmentos Retinianos/genética , Trombospondina 1/genética , Trombospondina 1/metabolismo
10.
Biochemistry ; 57(9): 1523-1532, 2018 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-29412660

RESUMEN

Macrophage migration inhibitory factor (MIF) is a pro-inflammatory and tumor-promoting cytokine that occurs in two redox-dependent immunologically distinct conformational isoforms. The disease-related structural isoform of MIF (oxMIF) can be specifically and predominantly detected in the circulation of patients with inflammatory diseases and in tumor tissue, whereas the ubiquitously expressed isoform of MIF (redMIF) is abundantly expressed in healthy and diseased subjects. In this article, we report that cysteine 81 within MIF serves as a "switch cysteine" for the conversion of redMIF to oxMIF. Modulating cysteine 81 by thiol reactive agents leads to significant structural rearrangements of the protein, resulting in a decreased ß-sheet content and an increased random coil content, but maintaining the trimeric quaternary structure. This conformational change in the MIF molecule enables binding of oxMIF-specific antibodies BaxB01 and BaxM159, which showed beneficial activity in animal models of inflammation and cancer. Crystal structure analysis of the MIF-derived EPCALCS peptide, bound in its oxMIF-like conformation by the Fab fragment of BaxB01, revealed that this peptide adopts a curved conformation, making the central thiol protein oxidoreductase motif competent to undergo disulfide shuffling. We conclude that redMIF might reflect a latent zymogenic form of MIF, and formation of oxMIF leads to a physiologically relevant, i.e., enzymatically active, state.


Asunto(s)
Cisteína/química , Cisteína/metabolismo , Oxidorreductasas Intramoleculares/química , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/química , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Especificidad de Anticuerpos , Dicroismo Circular , Cisteína/inmunología , Ensayo de Inmunoadsorción Enzimática , Mapeo Epitopo , Disulfuro de Glutatión/química , Disulfuro de Glutatión/metabolismo , Humanos , Oxidorreductasas Intramoleculares/inmunología , Factores Inhibidores de la Migración de Macrófagos/inmunología , Modelos Moleculares , Oxidación-Reducción , Conformación Proteica , Relación Estructura-Actividad
11.
J Biomol Struct Dyn ; 36(15): 4072-4084, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29210603

RESUMEN

A ten microsecond molecular dynamics simulation of a kallikrein-related peptidase 7 peptide complex revealed an unexpected change in binding mode. After more than two microseconds unrestrained sampling we observe a spontaneous transition of the binding pose including a 180° rotation around the P1 residue. Subsequently, the substrate peptide occupies the prime side region rather than the cognate non-prime side in a stable conformation. We characterize the unexpected binding mode in terms of contacts, solvent-accessible surface area, molecular interactions and energetic properties. We compare the new pose to inhibitor-bound structures of kallikreins with occupied prime side and find that a similar orientation is adopted. Finally, we apply in silico mutagenesis based on the alternative peptide binding position to explore the prime side specificity of kallikrein-related peptidase 7 and compare it to available experimental data. Our study provides the first microsecond time scale simulation data on a kallikrein protease and shows previously unexplored prime side interactions. Therefore, we expect our study to advance the rational design of inhibitors targeting kallikrein-related peptidase 7, an emerging drug target involved in several skin diseases as well as cancer.


Asunto(s)
Clorometilcetonas de Aminoácidos/química , Calicreínas/química , Simulación de Dinámica Molecular , Oligopéptidos/química , Inhibidores de Proteasas/química , Dominio Catalítico , Humanos , Calicreínas/antagonistas & inhibidores , Cinética , Ligandos , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Especificidad por Sustrato , Termodinámica
12.
Int J Mol Sci ; 17(12)2016 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-27898009

RESUMEN

Posttranslational modifications are an important feature of most proteases in higher organisms, such as the conversion of inactive zymogens into active proteases. To date, little information is available on the role of glycosylation and functional implications for secreted proteases. Besides a stabilizing effect and protection against proteolysis, several proteases show a significant influence of glycosylation on the catalytic activity. Glycans can alter the substrate recognition, the specificity and binding affinity, as well as the turnover rates. However, there is currently no known general pattern, since glycosylation can have both stimulating and inhibiting effects on activity. Thus, a comparative analysis of individual cases with sufficient enzyme kinetic and structural data is a first approach to describe mechanistic principles that govern the effects of glycosylation on the function of proteases. The understanding of glycan functions becomes highly significant in proteomic and glycomic studies, which demonstrated that cancer-associated proteases, such as kallikrein-related peptidase 3, exhibit strongly altered glycosylation patterns in pathological cases. Such findings can contribute to a variety of future biomedical applications.


Asunto(s)
Glicoproteínas/metabolismo , Péptido Hidrolasas/metabolismo , Glicoproteínas/química , Glicosilación , Cinética , Péptido Hidrolasas/química , Polisacáridos/química , Polisacáridos/metabolismo , Proteómica/métodos , Especificidad por Sustrato
13.
Biol Chem ; 397(12): 1251-1264, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27611765

RESUMEN

Although kallikrein-related peptidase 10 (KLK10) is expressed in a variety of human tissues and body fluids, knowledge of its physiological functions is fragmentary. Similarly, the pathophysiology of KLK10 in cancer is not well understood. In some cancer types, a role as tumor suppressor has been suggested, while in others elevated expression is associated with poor patient prognosis. Active human KLK10 exhibits a unique, three residue longer N-terminus with respect to other serine proteases and an extended 99-loop nearly as long as in tissue kallikrein KLK1. Crystal structures of recombinant ligand-free KLK10 and a Zn2+ bound form explain to some extent the mixed trypsin- and chymotrypsin-like substrate specificity. Zn2+-inhibition of KLK10 appears to be based on a unique mechanism, which involves direct binding and blocking of the catalytic triad. Since the disordered N-terminus and several loops adopt a zymogen-like conformation, the active protease conformation is very likely induced by interaction with the substrate, in particular at the S1 subsite and at the unusual Ser193 as part of the oxyanion hole. The KLK10 structures indicate that the N-terminus, the nearby 75-, 148-, and the 99-loops are connected in an allosteric network, which is present in other trypsin-like serine proteases with several variations.


Asunto(s)
Precursores Enzimáticos/antagonistas & inhibidores , Precursores Enzimáticos/química , Calicreínas/antagonistas & inhibidores , Calicreínas/química , Inhibidores de Proteasas/farmacología , Zinc/farmacología , Secuencia de Aminoácidos , Dominio Catalítico , Cristalografía por Rayos X , Precursores Enzimáticos/metabolismo , Humanos , Calicreínas/metabolismo , Modelos Moleculares , Inhibidores de Proteasas/metabolismo , Especificidad por Sustrato , Zinc/metabolismo
14.
Biol Chem ; 397(12): 1265-1276, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27483364

RESUMEN

Most members of the kallikrein-related peptidase family have been demonstrated to be dysregulated in ovarian cancer and modulate tumor growth, migration, invasion, and resistance to chemotherapy. In the present study, we assessed the mRNA expression levels of KLK6 and KLK8 by quantitative PCR in 100 patients with advanced serous ovarian cancer FIGO stage III/IV. A pronounced correlation between KLK6 and KLK8 mRNA expression (rs = 0.636, p < 0.001) was observed, indicating coordinate expression of both peptidases. No significant associations of clinical parameters with KLK6, KLK8, and a combined score KLK6+KLK8 were found. In univariate Cox regression analysis, elevated mRNA levels of KLK6 were significantly linked with shortened overall survival (OS) (hazard ratio [HR] = 2.07, p = 0.007). While KLK8 values were not associated with patients' outcome, high KLK6+KLK8 values were significantly associated with shorter progression-free survival (HR = 1.82, p = 0.047) and showed a trend towards significance in the case of OS (HR = 1.82, p = 0.053). Strikingly, in multivariable analysis, elevated KLK6 mRNA values, apart from residual tumor mass, remained an independent predictive marker for poor OS (HR = 2.33, p = 0.005). As KLK6 mRNA and protein levels correlate, KLK6 may represent an attractive therapeutic target for potent and specific inhibitors of its enzymatic activity.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Calicreínas/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Adulto , Anciano , Anciano de 80 o más Años , Supervivencia sin Enfermedad , Femenino , Humanos , Persona de Mediana Edad , Análisis Multivariante , Neoplasias Ováricas/enzimología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Adulto Joven
15.
J Biol Chem ; 291(2): 593-604, 2016 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-26582203

RESUMEN

Human kallikrein-related peptidase 2 (KLK2) is a key serine protease in semen liquefaction and prostate cancer together with KLK3/prostate-specific antigen. In order to decipher the function of its potential N-glycosylation site, we produced pro-KLK2 in Leishmania tarentolae cells and compared it with its non-glycosylated counterpart from Escherichia coli expression. Mass spectrometry revealed that Asn-95 carries a core glycan, consisting of two GlcNAc and three hexoses. Autocatalytic activation was retarded in glyco-pro-KLK2, whereas the activated glyco-form exhibited an increased proteolytic resistance. The specificity patterns obtained by the PICS (proteomic identification of protease cleavage sites) method are similar for both KLK2 variants, with a major preference for P1-Arg. However, glycosylation changes the enzymatic activity of KLK2 in a drastically substrate-dependent manner. Although glyco-KLK2 has a considerably lower catalytic efficiency than glycan-free KLK2 toward peptidic substrates with P2-Phe, the situation was reverted toward protein substrates, such as glyco-pro-KLK2 itself. These findings can be rationalized by the glycan-carrying 99-loop that prefers to cover the active site like a lid. By contrast, the non-glycosylated 99-loop seems to favor a wide open conformation, which mostly increases the apparent affinity for the substrates (i.e. by a reduction of Km). Also, the cleavage pattern and kinetics in autolytic inactivation of both KLK2 variants can be explained by a shift of the target sites due to the presence of the glycan. These striking effects of glycosylation pave the way to a deeper understanding of kallikrein-related peptidase biology and pathology.


Asunto(s)
Polisacáridos/metabolismo , Calicreínas de Tejido/química , Calicreínas de Tejido/metabolismo , Secuencia de Aminoácidos , Autólisis , Activación Enzimática , Fibronectinas/metabolismo , Glicosilación , Humanos , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Secundaria de Proteína , Proteolisis , Proteínas Recombinantes/aislamiento & purificación , Relación Estructura-Actividad , Especificidad por Sustrato , Factores de Tiempo
16.
J Biol Chem ; 289(49): 34267-83, 2014 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-25326387

RESUMEN

Human kallikrein-related peptidase 2 (KLK2) is a tryptic serine protease predominantly expressed in prostatic tissue and secreted into prostatic fluid, a major component of seminal fluid. Most likely it activates and complements chymotryptic KLK3 (prostate-specific antigen) in cleaving seminal clotting proteins, resulting in sperm liquefaction. KLK2 belongs to the "classical" KLKs 1-3, which share an extended 99- or kallikrein loop near their non-primed substrate binding site. Here, we report the 1.9 Å crystal structures of two KLK2-small molecule inhibitor complexes. In both structures discontinuous electron density for the 99-loop indicates that this loop is largely disordered. We provide evidence that the 99-loop is responsible for two biochemical peculiarities of KLK2, i.e. reversible inhibition by micromolar Zn(2+) concentrations and permanent inactivation by autocatalytic cleavage. Indeed, several 99-loop mutants of KLK2 displayed an altered susceptibility to Zn(2+), which located the Zn(2+) binding site at the 99-loop/active site interface. In addition, we identified an autolysis site between residues 95e and 95f in the 99-loop, whose elimination prevented the mature enzyme from limited autolysis and irreversible inactivation. An exhaustive comparison of KLK2 with related structures revealed that in the KLK family the 99-, 148-, and 220-loop exist in open and closed conformations, allowing or preventing substrate access, which extends the concept of conformational selection in trypsin-related proteases. Taken together, our novel biochemical and structural data on KLK2 identify its 99-loop as a key player in activity regulation.


Asunto(s)
Calicreínas/química , Zinc/química , Secuencia de Aminoácidos , Cationes Bivalentes , Cristalografía por Rayos X , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Calicreínas/genética , Calicreínas/metabolismo , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Relación Estructura-Actividad , Zinc/metabolismo
17.
Biol Chem ; 395(9): 959-76, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25153382

RESUMEN

Most kallikrein-related peptidases (KLKs) are N-glycosylated with N-acetylglucosamine2-mannose9 units at Asn-Xaa-Ser/Thr sequons during protein synthesis and translocation into the endoplasmic reticulum. These N-glycans are modified in the Golgi machinery, where additional O-glycosylation at Ser and Thr takes place, before exocytotic release of the KLKs into the extracellular space. Sequons are present in all 15 members of the KLKs and comparative studies for KLKs from natural and recombinant sources elucidated some aspects of glycosylation. Although glycosylation of mammalian KLKs 1, 3, 4, 6, and 8 has been analyzed in great detail, e.g., by crystal structures, the respective function remains largely unclear. In some cases, altered enzymatic activity was observed for KLKs upon glycosylation. Remarkably, for KLK3/PSA, changes in the glycosylation pattern were observed in samples of benign prostatic hyperplasia and prostate cancer with respect to healthy individuals. Potential functions of KLK glycosylation in structural stabilization, protection against degradation, and activity modulation of substrate specificity can be deduced from a comparison with other glycosylated proteins and their regulation. According to the new concept of protein sectors, glycosylation distant from the active site might significantly influence the activity of proteases. Novel pharmacological approaches can exploit engineered glycans in the therapeutical context.


Asunto(s)
Calicreínas/metabolismo , Polisacáridos/metabolismo , Secuencia de Aminoácidos , Animales , Glicoproteínas/metabolismo , Glicosilación , Humanos , Calicreínas/química , Datos de Secuencia Molecular , Polisacáridos/química , Especificidad por Sustrato
18.
Crit Rev Clin Lab Sci ; 51(2): 63-84, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24490956

RESUMEN

Gynecological cancers, including malignant tumors of the ovaries, the endometrium and the cervix, account for approximately 10% of tumor-associated deaths in women of the Western world. For screening, diagnosis, prognosis, and therapy response prediction, the group of enzymes known as serine (Ser-)proteases show great promise as biomarkers. In the present review, following a summary of the clinical facts regarding malignant tumors of the ovaries, the endometrium and the cervix, and characterization of the most important Ser-proteases, we thoroughly review the current state of knowledge relating to the use of proteases as biomarkers of the most frequent gynecological cancers. Within the Ser-protease group, the kallikrein-related peptidase (KLK) family, which encompasses a subgroup of 15 members, holds particular promise, with some acting via a tumor-promoting mechanism and others behaving as protective factors. Further, the urokinase-type plasminogen activator (uPA) and its inhibitor PAI-1 (plasminogen activator inhibitor-1) seem to play an unfavorable role in gynecological tumors, while down-regulation of high-temperature requirement proteins A 1, 2 and 3 (HtrA1,2,3) is associated with malignant disease and cancer progression. Expression/activity levels of other Ser-proteases, including the type II transmembrane Ser-proteases (TTSPs) matriptase, hepsin (TMPRSS1), and the hepsin-related protease (TMPRSS3), as well as the glycosyl-phosphatidylinositol (GPI)-anchored Ser-proteases prostasin and testisin, may be of clinical relevance in gynecological cancers. In conclusion, proteases are a rich source of biomarkers of gynecological cancer, though the enzymes' exact roles and functions merit further investigation.


Asunto(s)
Biomarcadores de Tumor , Neoplasias de los Genitales Femeninos , Calicreínas , Femenino , Humanos , Serina Proteasas
19.
J Biotechnol ; 168(4): 421-5, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24184090

RESUMEN

Enterokinase, a two-chain duodenal serine protease, activates trypsinogen by removing its N-terminal propeptide. Due to a clean cut after the non-primed site recognition sequence, the enterokinase light chain is frequently employed in biotechnology to separate N-terminal affinity tags from target proteins with authentic N-termini. In order to obtain large quantities of this protease, we adapted an in vitro folding protocol for a pentahistidine-tagged triple mutant of the bovine enterokinase light chain. The purified, highly active enzyme successfully processed recombinant target proteins, while the pentahistidine-tag facilitated post-cleavage removal. Hence, we conclude that producing enterokinase in one's own laboratory is an efficient alternative to the commercial enzyme.


Asunto(s)
Enteropeptidasa/química , Enteropeptidasa/genética , Histidina/química , Ingeniería de Proteínas , Animales , Bovinos , Clonación Molecular , Enteropeptidasa/biosíntesis , Escherichia coli/genética , Cuerpos de Inclusión/enzimología , Pliegue de Proteína , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética
20.
Biochimie ; 92(11): 1546-67, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20615447

RESUMEN

Including the true tissue kallikrein KLK1, kallikrein-related peptidases (KLKs) represent a family of fifteen mammalian serine proteases. While the physiological roles of several KLKs have been at least partially elucidated, their activation and regulation remain largely unclear. This obscurity may be related to the fact that a given KLK fulfills many different tasks in diverse fetal and adult tissues, and consequently, the timescale of some of their physiological actions varies significantly. To date, a variety of endogenous inhibitors that target distinct KLKs have been identified. Among them are the attenuating Zn(2+) ions, active site-directed proteinaceous inhibitors, such as serpins and the Kazal-type inhibitors, or the huge, unspecific compartment forming α(2)-macroglobulin. Failure of these inhibitory systems can lead to certain pathophysiological conditions. One of the most prominent examples is the Netherton syndrome, which is caused by dysfunctional domains of the Kazal-type inhibitor LEKTI-1 which fail to appropriately regulate KLKs in the skin. Small synthetic inhibitory compounds and natural polypeptidic exogenous inhibitors have been widely employed to characterize the activity and substrate specificity of KLKs and to further investigate their structures and biophysical properties. Overall, this knowledge leads not only to a better understanding of the physiological tasks of KLKs, but is also a strong fundament for the synthesis of small compound drugs and engineered biomolecules for pharmaceutical approaches. In several types of cancer, KLKs have been found to be overexpressed, which makes them clinically relevant biomarkers for prognosis and monitoring. Thus, down regulation of excessive KLK activity in cancer and in skin diseases by small inhibitor compounds may represent attractive therapeutical approaches.


Asunto(s)
Productos Biológicos/farmacología , Calicreínas/antagonistas & inhibidores , Inhibidores de Proteasas/farmacología , Secuencia de Aminoácidos , Animales , Productos Biológicos/síntesis química , Productos Biológicos/química , Descubrimiento de Drogas , Humanos , Calicreínas/química , Datos de Secuencia Molecular , Inhibidores de Proteasas/síntesis química , Inhibidores de Proteasas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA