Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 291
Filtrar
Más filtros

Intervalo de año de publicación
1.
Dokl Biochem Biophys ; 509(1): 78-80, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37340298

RESUMEN

On the basis of known published data, six peptide sequences were selected that are potentially capable of being rapidly cleaved by the endosomal protease cathepsin B. For comparison, the cleavage of common linker sequences, polyglycine and polyglycine-serine, by cathepsin B was also studied. Different ends of these peptides were labeled with sulfoCyanine3 and sulfoCyanine5 fluorescent dyes, between which Förster resonant energy transfer (FRET) is possible. The kinetics of cleavage of peptides by cathepsin B was studied on a multimodal plate reader by FRET signal reduction. FKFL and FRRG cleavage sites have been shown to be the most suitable for potential use in various drug delivery systems. These sites are much more efficiently cleaved under slightly acidic conditions of endosomes than at neutral extracellular pH.


Asunto(s)
Aminoácidos , Catepsina B , Catepsina B/química , Catepsina B/metabolismo , Aminoácidos/metabolismo , Cinética , Péptidos/química , Endosomas/metabolismo , Sistemas de Liberación de Medicamentos
2.
Biochemistry ; 61(17): 1904-1914, 2022 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-35981509

RESUMEN

Cathepsin B is a lysosomal protease that participates in protein degradation. However, cathepsin B is also active under neutral pH conditions of the cytosol, nuclei, and extracellular locations. The dipeptidyl carboxypeptidase (DPCP) activity of cathepsin B, assayed with the Abz-GIVR↓AK(Dnp)-OH substrate, has been reported to display an acidic pH optimum. In contrast, the endopeptidase activity, monitored with Z-RR-↓AMC, has a neutral pH optimum. These observations raise the question of whether other substrates can demonstrate cathepsin B DPCP activity at neutral pH and endopeptidase activity at acidic pH. To address this question, global cleavage profiling of cathepsin B with a diverse peptide library was conducted under acidic and neutral pH conditions. Results revealed that cathepsin B has (1) major DPCP activity and modest endopeptidase activity under both acidic and neutral pH conditions and (2) distinct pH-dependent amino acid preferences adjacent to cleavage sites for both DPCP and endopeptidase activities. The pH-dependent cleavage preferences were utilized to design a new Abz-GnVR↓AK(Dnp)-OH DPCP substrate, with norleucine (n) at the P3 position, having improved DPCP activity of cathepsin B at neutral pH compared to the original Abz-GIVR↓AK(Dnp)-OH substrate. The new Z-VR-AMC and Z-ER-AMC substrates displayed improved endopeptidase activity at acidic pH compared to the original Z-RR-AMC. These findings illustrate the new concept that cathepsin B possesses DPCP and endopeptidase activities at both acidic and neutral pH values. These results advance understanding of the pH-dependent cleavage properties of the dual DPCP and endopeptidase activities of cathepsin B that function under different cellular pH conditions.


Asunto(s)
Catepsina B , Catepsinas , Catepsina B/química , Catepsinas/metabolismo , Endopeptidasas , Concentración de Iones de Hidrógeno , Hidrólisis , Proteolisis , Especificidad por Sustrato
3.
Int J Mol Sci ; 23(9)2022 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-35563462

RESUMEN

The human gonadotropin releasing hormone (GnRH-I) and its sea lamprey analogue GnRH-III specifically bind to GnRH receptors on cancer cells and can be used as targeting moieties for targeted tumor therapy. Considering that the selective release of drugs in cancer cells is of high relevance, we were encouraged to develop cleavable, self-immolative GnRH-III-drug conjugates which consist of a p-aminobenzyloxycarbonlyl (PABC) spacer between a cathepsin B-cleavable dipeptide (Val-Ala, Val-Cit) and the classical anticancer drugs daunorubicin (Dau) and paclitaxel (PTX). Alongside these compounds, non-cleavable GnRH-III-drug conjugates were also synthesized, and all compounds were analyzed for their antiproliferative activity. The cleavable GnRH-III bioconjugates revealed a growth inhibitory effect on GnRH receptor-expressing A2780 ovarian cancer cells, while their activity was reduced on Panc-1 pancreatic cancer cells exhibiting a lower GnRH receptor level. Moreover, the antiproliferative activity of the non-cleavable counterparts was strongly reduced. Additionally, the efficient cleavage of the Val-Ala linker and the subsequent release of the drugs could be verified by lysosomal degradation studies, while radioligand binding studies ensured that the GnRH-III-drug conjugates bound to the GnRH receptor with high affinity. Our results underline the high value of GnRH-III-based homing devices and the application of cathepsin B-cleavable linker systems for the development of small molecule drug conjugates (SMDCs).


Asunto(s)
Hormona Liberadora de Gonadotropina , Terapia Molecular Dirigida , Neoplasias Ováricas , Receptores LHRH , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/química , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Catepsina B/química , Catepsina B/uso terapéutico , Línea Celular Tumoral , Daunorrubicina/química , Daunorrubicina/uso terapéutico , Femenino , Hormona Liberadora de Gonadotropina/uso terapéutico , Humanos , Terapia Molecular Dirigida/métodos , Paclitaxel/química , Paclitaxel/uso terapéutico , Petromyzon , Ácido Pirrolidona Carboxílico/análogos & derivados , Ácido Pirrolidona Carboxílico/uso terapéutico , Receptores LHRH/uso terapéutico
4.
PLoS Comput Biol ; 16(12): e1008461, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33290397

RESUMEN

The entry of SARS-CoV-2 into target cells requires the activation of its surface spike protein, S, by host proteases. The host serine protease TMPRSS2 and cysteine proteases Cathepsin B/L can activate S, making two independent entry pathways accessible to SARS-CoV-2. Blocking the proteases prevents SARS-CoV-2 entry in vitro. This blockade may be achieved in vivo through 'repurposing' drugs, a potential treatment option for COVID-19 that is now in clinical trials. Here, we found, surprisingly, that drugs targeting the two pathways, although independent, could display strong synergy in blocking virus entry. We predicted this synergy first using a mathematical model of SARS-CoV-2 entry and dynamics in vitro. The model considered the two pathways explicitly, let the entry efficiency through a pathway depend on the corresponding protease expression level, which varied across cells, and let inhibitors compromise the efficiency in a dose-dependent manner. The synergy predicted was novel and arose from effects of the drugs at both the single cell and the cell population levels. Validating our predictions, available in vitro data on SARS-CoV-2 and SARS-CoV entry displayed this synergy. Further, analysing the data using our model, we estimated the relative usage of the two pathways and found it to vary widely across cell lines, suggesting that targeting both pathways in vivo may be important and synergistic given the broad tissue tropism of SARS-CoV-2. Our findings provide insights into SARS-CoV-2 entry into target cells and may help improve the deployability of drug combinations targeting host proteases required for the entry.


Asunto(s)
Catepsina B/química , Catepsina L/química , SARS-CoV-2/fisiología , Serina Endopeptidasas/química , Internalización del Virus/efectos de los fármacos , Animales , Chlorocebus aethiops , Reposicionamiento de Medicamentos , Humanos , Modelos Teóricos , Unión Proteica , Glicoproteína de la Espiga del Coronavirus/genética , Células Vero , Virión , Tratamiento Farmacológico de COVID-19
5.
J Comput Aided Mol Des ; 35(2): 223-244, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33458809

RESUMEN

Here we present WIDOCK, a virtual screening protocol that supports the selection of diverse electrophiles as covalent inhibitors by incorporating ligand reactivity towards cysteine residues into AutoDock4. WIDOCK applies the reactive docking method (Backus et al. in Nature 534:570-574, 2016) and extends it into a virtual screening tool by introducing facile experimental or computational parametrization and a ligand focused evaluation scheme together with a retrospective and prospective validation against various therapeutically relevant targets. Parameters accounting for ligand reactivity are derived from experimental reaction kinetic data or alternatively from computed reaction barriers. The performance of this docking protocol was first evaluated by investigating compound series with diverse warhead chemotypes against KRASG12C, MurA and cathepsin B. In addition, WIDOCK was challenged on larger electrophilic libraries screened against OTUB2 and NUDT7. These retrospective analyses showed high sensitivity in retrieving experimental actives, by also leading to superior ROC curves, AUC values and better enrichments than the standard covalent docking tool available in AutoDock4 when compound collections with diverse warheads were investigated. Finally, we applied WIDOCK for the prospective identification of covalent human MAO-A inhibitors acting via a new mechanism by binding to Cys323. The inhibitory activity of several predicted compounds was experimentally confirmed and the labelling of Cys323 was proved by subsequent MS/MS measurements. These findings demonstrate the usefulness of WIDOCK as a warhead-sensitive, covalent virtual screening protocol.


Asunto(s)
Transferasas Alquil y Aril/química , Catepsina B/química , Inhibidores Enzimáticos/química , Proteínas Proto-Oncogénicas p21(ras)/química , Secuencia de Aminoácidos , Sitios de Unión , Cisteína/química , Glutatión/química , Ligandos , Simulación del Acoplamiento Molecular , Unión Proteica , Conformación Proteica , Programas Informáticos , Relación Estructura-Actividad
6.
Bioorg Chem ; 117: 105463, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34753058

RESUMEN

Human cathepsin B is a cysteine-dependent protease whose roles in both normal and diseased cellular states remain yet to be fully delineated. This is primarily due to overlapping substrate specificities and lack of unambiguously annotated physiological functions. In this work, a selective, cell-permeable, clickable and tagless small molecule cathepsin B probe, KDA-1, is developed and kinetically characterized. KDA-1 selectively targets active site Cys25 residue of cathepsin B for labeling and can detect active cellular cathepsin B in proteomes derived from live human MDA-MB-231 breast cancer cells and HEK293 cells. It is anticipated that KDA-1 probe will find suitable applications in functional proteomics involving human cathepsin B enzyme.


Asunto(s)
Catepsina B/química , Sondas Moleculares/química , Catepsina B/genética , Línea Celular , Relación Dosis-Respuesta a Droga , Humanos , Sondas Moleculares/síntesis química , Estructura Molecular , Relación Estructura-Actividad
7.
Angew Chem Int Ed Engl ; 60(22): 12341-12345, 2021 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-33759310

RESUMEN

Protein assemblies can be designed for development of nano-bio materials. This has been achieved by modulating protein-protein interactions. However, fabrication of highly ordered protein assemblies remains challenging. Protein crystals, which have highly ordered arrangements of protein molecules, provide useful source matrices for synthesizing artificial protein assemblies. Here, we describe construction of a supramolecular filament structure by engineering covalent and non-covalent interactions in a protein crystal. Performing in-cell crystallization of Trypanosoma brucei cysteine protease cathepsin B (TbCatB), we achieved a precise arrangement of protein molecules while suppressing random aggregation due to disulfide bonds. We succeeded in synthesizing bundled filament from the crystals by autoxidation of cysteinyl thiols after the isolation of the crystals from living cells.


Asunto(s)
Catepsina B/química , Citoesqueleto/metabolismo , Proteínas Protozoarias/química , Catepsina B/genética , Catepsina B/metabolismo , Cristalización , Citoesqueleto/química , Concentración de Iones de Hidrógeno , Mutagénesis , Estructura Terciaria de Proteína , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Trypanosoma brucei brucei/metabolismo
8.
Biochemistry ; 59(14): 1420-1427, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32212642

RESUMEN

Cathepsin B (CTSB) is an abundant cysteine protease that functions in both endolysosomal compartments and extracellular regions. A considerable number of preclinical and clinical studies indicate that CTSB is implicated in many human diseases. Expression levels and activity of CTSB significantly correlate with disease progression and severity. Current inhibitors of CTSB are lack of adequate specificity and pharmacological activities. Through structure-guided rational design, we hereby designed and generated a humanized antibody inhibitor targeting human CTSB. This was achieved by genetically fusing the propeptide of procathepsin B, a naturally occurring inhibitor of CTSB, into heavy chain complementarity-determining region 3 (CDR3H) of Herceptin that is used in the clinic for the treatment of breast cancer. The resulting antibody-propeptide fusion displayed high specificity for inhibiting CTSB proteolytic activity at nanomolar levels. Pharmacokinetic studies in mice revealed a plasma half-life of approximately 42 h for this anti-CTSB antibody inhibitor, comparable to that of the parental Herceptin scaffold. This study demonstrates a new approach for the efficient generation of humanized antibody inhibitors with high potency and specificity for human CTSB, which may be extended to develop antibody inhibitors against other disease relevant cathepsin proteases.


Asunto(s)
Anticuerpos/química , Catepsina B/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Animales , Anticuerpos/administración & dosificación , Anticuerpos/genética , Anticuerpos/metabolismo , Catepsina B/química , Catepsina B/genética , Catepsina B/metabolismo , Diseño de Fármacos , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/metabolismo , Femenino , Humanos , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Dominios Proteicos
9.
Drug Metab Dispos ; 47(8): 884-889, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31072822

RESUMEN

Carfilzomib (CFZ) is a proteasome inhibitor used for oncology indications including treating multiple myeloma. CFZ is a potent cytotoxic agent with an IC50 value in the nanomolar range in various cancer cell lines and was considered as a potential payload for antibody drug conjugates (ADCs); however, the conjugated CFZ to anti-CD22 or anti-HER2 antibody totally abolishes the in vitro potency. This was a surprise since with other payloads such as monomethyl auristatin E (MMAE), where potent antiproliferation efficacy was retained as MMAE alone or as a payload in an ADC. Further investigations were conducted using CFZ alone, CFZ with a linker, and CFZ-ADC with tissue matrices including lysosomal enzymes. With CFZ linked to the ADC, cathepsin B (a lysosomal enzyme) was efficient in liberating CFZ from the ADC by cleavage of the valine-citrulline linker. At the same time, the liberated CFZ in the lysosome was inactivated due to further metabolism by lysosomal enzymes. The products from epoxide and amide hydrolysis were identified from these incubations. These results suggested that the CFZ-ADC upon uptake and internalization specifically delivers CFZ payload to the lysosomes, where CFZ was inactivated. On the other hand, CFZ by itself is not as vulnerable and could reach its target. Therefore, lysosomal stability is an important criterion in the selection of a payload for making the next generation of potent ADC therapeutics.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Antineoplásicos/farmacocinética , Inmunoconjugados/farmacocinética , Lisosomas/enzimología , Oligopéptidos/farmacocinética , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/química , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Catepsina B/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Estudios de Factibilidad , Humanos , Inmunoconjugados/administración & dosificación , Inmunoconjugados/química , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Oligopéptidos/administración & dosificación , Oligopéptidos/química , Receptor ErbB-2/antagonistas & inhibidores , Lectina 2 Similar a Ig de Unión al Ácido Siálico/antagonistas & inhibidores
10.
Biomacromolecules ; 20(9): 3547-3556, 2019 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-31381303

RESUMEN

Detection and removal of preneoplastic tumors is crucial for successful colorectal cancer (CRC) therapy. Here we describe the design of a Cathepsin B (CB)-activated polymeric probe, P-(GGFLGK-IR783), for imaging CRC tumors established by intrarectal or subcutaneous (s.c.) implantation of human colon cancer cells (SW-480 and HT-29) in mice. Multiple copies of the near-infrared fluorescent (NIRF) dye IR783 were attached to a single HPMA copolymer backbone via a CB-cleavable linker (GFLG), and the influence of the dye loading on the fluorescence quenching and activation by CB was assessed in vitro, ex vivo, and in vivo. The optimal dose and dosing regimen of P-(GGFLGK-IR783) for colonic tumor detection was determined. Increasing the IR783 loading in the copolymer from 2.5 to 20 mol % resulted in quenching of the fluorescence signal that was activated in vitro by the action of CB from different origins. Following intravenous administration, P-(GGFLGK-IR783)7.5% preferentially accumulated in intrarectal and s.c. implanted tumors, allowing tumor visualization after 4 h and even 48 h postadministration. Activation of P-(GGFLGK-IR783)7.5% by CB was clearly detected in s.c. implanted tumors, revealing about a 4-fold increase in the fluorescence signal in tumors vs healthy colon tissue. The probe containing the CB-cleavable linker produced higher fluorescence signal intensity in tumors, relative to the noncleavable probe. These results indicate that P-(GGFLGK-IR783)7.5% may aid in detecting CRC tumors and can help to guide selective removal of polyps during colonoscopic procedures.


Asunto(s)
Catepsina B/farmacología , Neoplasias Colorrectales/diagnóstico por imagen , Colorantes Fluorescentes/farmacología , Imagen Molecular , Animales , Catepsina B/química , Neoplasias Colorrectales/patología , Colorantes Fluorescentes/química , Células HT29 , Xenoinjertos , Humanos , Ratones , Polímeros/química , Polímeros/farmacología , Espectroscopía Infrarroja Corta
11.
Inorg Chem ; 58(1): 294-306, 2019 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-30475597

RESUMEN

Decoding the interaction between coordination compounds and proteins is of fundamental importance in biology, pharmacy, and medicine. In this context, protein- ligand docking represents a particularly interesting asset to predict how small compounds could interact with biomolecules, but to date, very little information is available to adapt these methodologies to metal-containing ligands. Here, we assessed the predictive capability of a metal-compatible parameter set for the docking program GOLD for metallo ligands with multiple vacant sites and different geometries. The study first presents a benchmark of 25 well-characterized X-ray metallo ligand-protein adducts. In 100% of the cases, the docking solutions are superimposable to the X-ray determination, and in 92% the value of the root-mean-square deviation between the experimental and calculated structures is lower than 1.5 Å. After the validation step, we applied these methods to five case studies for the prediction of the binding of pharmacological active metal species to proteins: (i) the anticancer copper(II) complex [CuII(Br)(2-hydroxy-1-naphthaldehyde benzoyl hydrazine)(indazole)] to human serum albumin (HSA); (ii) one of the active species of antidiabetic and antitumor vanadium compounds, VIVO2+ ion, to carboxypeptidase; (iii) the antiarthritic species [AuI(PEt3)]+ to HSA; (iv) the antitumor oxaliplatin to ubiquitin; (v) the antitumor ruthenium(II) compound RAPTA-PentaOH to cathepsin B. The calculations suggested that the binding modes are in good agreement with the partial information retrieved from spectroscopic and spectrometric analysis and allowed us, in certain cases, to propose additional hypotheses. This method is an important update in protein-metallo ligand docking, which could have a wide field of application, from biology and inorganic biochemistry to medicinal chemistry and pharmacology.


Asunto(s)
Carboxipeptidasas/metabolismo , Catepsina B/metabolismo , Complejos de Coordinación/metabolismo , Albúmina Sérica Humana/metabolismo , Ubiquitina/metabolismo , Sitios de Unión , Carboxipeptidasas/química , Catepsina B/química , Complejos de Coordinación/química , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Unión Proteica , Albúmina Sérica Humana/química , Ubiquitina/química
12.
Bioorg Med Chem ; 27(1): 1-15, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30473362

RESUMEN

The potential of papain-like cysteine proteases, such as cathepsin B, as drug discovery targets for systemic human diseases has prevailed over the past years. The development of potent and selective low-molecular cathepsin B inhibitors relies on the detailed expertise on preferred amino acid and inhibitor residues interacting with the corresponding specificity pockets of cathepsin B. Such knowledge might be obtained by mapping the active site of the protease with combinatorial libraries of peptidic substrates and peptidomimetic inhibitors. This review, for the first time, summarizes a wide spectrum of active site mapping approaches. It considers relevant X-ray crystallographic data and discloses propensities towards favorable protein-ligand interactions in case of the therapeutically relevant protease cathepsin B.


Asunto(s)
Catepsina B/química , Inhibidores de Cisteína Proteinasa/química , Péptidos/química , Animales , Dominio Catalítico , Cristalografía por Rayos X , Humanos , Cinética , Ligandos , Especificidad por Sustrato
13.
J Biol Chem ; 292(51): 21071-21082, 2017 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-29046353

RESUMEN

ß-Amyloid (Aß) aggregation is thought to initiate a cascade of neurodegenerative events in Alzheimer's disease (AD). Much effort is underway to develop strategies to reduce Aß concentration or inhibit aggregation. Cathepsin B (CatB) proteolytically degrades Aß into non-aggregating fragments but is potently inhibited by cystatin C (CysC). It has been suggested that decreasing CysC would facilitate Aß clearance by relieving CatB inhibition. However, CysC binds Aß and inhibits Aß aggregation, suggesting that an intervention that increases CysC would prevent Aß aggregation. Both approaches have been tested in animal models, yielding contradictory results, possibly because of the opposing influences of CysC on Aß degradation versus aggregation. Here, we sought to develop a model that quantitatively predicts the effects of CysC and CatB on Aß aggregation. Aß aggregation kinetics in the absence of CatB or CysC was measured. The rate constant for Aß degradation by CatB and the equilibrium constant for binding of CysC to Aß were determined. We derived a mathematical model that combines material balances and kinetic rate equations. The model accurately predicted Aß aggregation kinetics at various CatB and CysC concentrations. We derived approximate expressions for the half-times of degradation and aggregation and show that their ratio can be used to estimate, at any given Aß, CatB, or CysC concentration, whether Aß aggregation or degradation will result. Our results may be useful for designing experiments and interpreting results from investigations of manipulation of CysC concentration as an AD therapy.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Catepsina B/metabolismo , Cistatina C/metabolismo , Modelos Moleculares , Proteínas del Tejido Nervioso/metabolismo , Fragmentos de Péptidos/metabolismo , Agregación Patológica de Proteínas/metabolismo , Algoritmos , Sustitución de Aminoácidos , Péptidos beta-Amiloides/química , Catepsina B/química , Cistatina C/química , Cistatina C/genética , Transferencia Resonante de Energía de Fluorescencia , Humanos , Cinética , Mutagénesis Sitio-Dirigida , Mutación , Proteínas del Tejido Nervioso/química , Concentración Osmolar , Fragmentos de Péptidos/química , Dominios y Motivos de Interacción de Proteínas , Proteolisis , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Reproducibilidad de los Resultados , Solubilidad
14.
Biol Chem ; 399(10): 1223-1235, 2018 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-29924726

RESUMEN

The genome of the model plant Arabidopsis thaliana encodes three paralogues of the papain-like cysteine proteinase cathepsin B (AtCathB1, AtCathB2 and AtCathB3), whose individual functions are still largely unknown. Here we show that a mutated splice site causes severe truncations of the AtCathB1 polypeptide, rendering it catalytically incompetent. By contrast, AtCathB2 and AtCathB3 are effective proteases which display comparable hydrolytic properties and share most of their substrate specificities. Site-directed mutagenesis experiments demonstrated that a single amino acid substitution (Gly336→Glu) is sufficient to confer AtCathB2 with the capacity to tolerate arginine in its specificity-determining S2 subsite, which is otherwise a hallmark of AtCathB3-mediated cleavages. A degradomics approach utilizing proteome-derived peptide libraries revealed that both enzymes are capable of acting as endopeptidases and exopeptidases, releasing dipeptides from the C-termini of substrates. Mutation of the carboxydipeptidase determinant His207 also affected the activity of AtCathB2 towards non-exopeptidase substrates, highlighting mechanistic differences between plant and human cathepsin B. This was also noted in molecular modeling studies which indicate that the occluding loop defining the dual enzymatic character of cathepsin B does not obstruct the active-site cleft of AtCathB2 to the same extent as in its mammalian orthologues.


Asunto(s)
Arabidopsis/enzimología , Carboxipeptidasas/metabolismo , Catepsina B/metabolismo , Endopeptidasas/metabolismo , Animales , Carboxipeptidasas/química , Carboxipeptidasas/genética , Catepsina B/química , Catepsina B/genética , Clonación Molecular , Endopeptidasas/química , Endopeptidasas/genética , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Hojas de la Planta/enzimología , Reacción en Cadena en Tiempo Real de la Polimerasa , Spodoptera/citología , Spodoptera/genética
15.
Mol Pharm ; 15(10): 4668-4676, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30179491

RESUMEN

Gold nanoparticles are commonly used for medical applications such as drug delivery and as therapeutic and diagnostic materials because of their unique properties. In this study, we prepared docetaxel (DTX)-loaded hyaluronic acid-cleavable-peptide-gold nanoparticles for the treatment of cancer by selectively delivering DTX into the tumor and, thus, enhancing the therapeutic effect of DTX; further, we determined synergistic effects of the nanoparticles using laser treatment. The DTX-loaded hyaluronic acid-cleavable-peptide-gold nanoparticles prepared in this study had an average size of 75 nm and negative surface charge. The nanoparticles revealed greater cytotoxicity and higher tumor suppression efficacy in tumor models than free DTX under near-infrared laser irradiation. Therefore, the nanoparticle formulation prepared in this study could be utilized for targeted drug delivery and in combination with other cancer therapies.


Asunto(s)
Catepsina B/química , Docetaxel/química , Oro/química , Ácido Hialurónico/química , Nanopartículas del Metal/química , Péptidos/química , Polímeros/química , Animales , Sistemas de Liberación de Medicamentos/métodos , Humanos
16.
Mol Pharm ; 15(10): 4568-4576, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30107748

RESUMEN

PEGylation typically improves the systemic exposure and tumor biodistribution of polymeric drug delivery systems, but may also restrict enzyme access to peptide-based drug linkers. The impact of dendrimer generation (G4 vs G5) and PEG length (570 vs 1100 Da) on the pharmacokinetics, tumor biodistribution, drug release kinetics, and anticancer activity of a series of PEGylated polylysine dendrimers conjugated with doxorubicin via a cathepsin-B cleavable valine-citrulline linker was therefore investigated in rodents. Although the smallest G4 PEG570 dendrimer showed the most efficient cathepsin-mediated doxorubicin release, systemic exposure and tumor uptake were limited. The largest G5 PEG1100 dendrimer showed good tumor uptake and retention but restricted drug liberation and therefore limited anticancer activity. Superior anticancer activity was achieved using an intermediate sized dendrimer that showed better drug release kinetics, systemic exposure, tumor uptake, and retention. The data suggest that balancing PEG molecular weight and dendrimer size is critical when designing chemotherapeutic dendrimers.


Asunto(s)
Catepsinas/química , Dendrímeros/química , Doxorrubicina/química , Polilisina/química , Células A549 , Animales , Catepsina B/química , Sistemas de Liberación de Medicamentos , Liberación de Fármacos , Humanos , Masculino , Polietilenglicoles/química , Ratas
17.
Mar Drugs ; 16(2)2018 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-29466301

RESUMEN

Three new polyketides, lactomycins A (1)-C (3), were isolated from the culture broth of a marine-derived Streptomyces sp. ACT232 as cathepsin B inhibitors. Their structures were determined by a combination of NMR and MS data analyses to be the dephosphorylated derivatives of a phoslactomycin class of metabolites. Lactomycins exhibited cathepsin B inhibitory activity (IC50 0.8 to 4.5 µg/mL). Even though the biosynthetic gene clusters found in the genome of the current strain have high similarity to those of phoslactomycin, neither phoslactomycins nor leustroducsins were detected by LC-MS analyses of the crude extract.


Asunto(s)
Catepsina B/química , Streptomyces/química , Antifúngicos/química , Indoles/química , Espectroscopía de Resonancia Magnética/métodos , Naftoquinonas/química , Policétidos/química
18.
Mol Pharm ; 14(5): 1450-1459, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28277671

RESUMEN

Peptides derived from the third Bcl-2 homology domain (BH3) renormalize apoptotic signaling by antagonizing prosurvival Bcl-2 family members. These potential peptide drugs exhibit therapeutic activities but are limited by barriers including short circulation half-lives and poor penetration into cells. A diblock polymeric micelle carrier for the BIM BH3 peptide was recently described that demonstrated antitumor activity in a B-cell lymphoma xenograft model [Berguig et al., Mol. Ther. 2015, 23, 907-917]. However, the disulfide linkage used to conjugate the BIM peptide was shown to have nonoptimal blood stability. Here we describe a peptide macromonomer composed of BIM capped with a four amino acid cathepsin B substrate (FKFL) that possesses high blood stability and is cleaved to release the drug inside of target cells. Employing RAFT polymerization, the peptide macromonomer was directly integrated into a multifunctional diblock copolymer tailored for peptide delivery. The first polymer block was made as a macro-chain transfer agent (CTA) and composed of a pH-responsive endosomolytic formulation of N,N-diethylaminoethyl methacrylate (DEAEMA) and butyl methacrylate (BMA). The second polymer block was a copolymer of the peptide and polyethylene glycol methacrylate (PEGMA). PEGMA monomers of two sizes were investigated (300 Da and 950 Da). Protein gel analysis, high performance liquid chromatography, and coupled mass spectrometry (MS) showed that incubation with cathepsin B specifically cleaved the FKFL linker and released active BIM peptide with PEGMA300 but not with PEGMA950. MALDI-TOF MS showed that incubation of the peptide monomers alone in human serum resulted in partial cleavage at the FKFL linker after 12 h. However, formulation of the peptides into polymers protected against serum-mediated peptide degradation. Dynamic light scattering (DLS) demonstrated pH-dependent micelle disassembly (25 nm polymer micelles at pH 7.4 versus 6 nm unimers at pH 6.6), and a red blood cell lysis assay showed a corresponding increase in membrane destabilizing activity (<1% lysis at pH 7.4 versus 95% lysis at pH 6.6). The full carrier-drug system successfully induced apoptosis in SKOV3 ovarian cancer cells in a dose-dependent manner, in comparison to a control polymer containing a scrambled BIM peptide sequence. Mechanistic analysis verified target-dependent activation of caspase 3/7 activity (8.1-fold increase), and positive annexin V staining (72% increase). The increased blood stability of this enzyme-cleavable peptide polymer design, together with the direct polymerization approach that eliminated postsynthetic conjugation steps, suggests that this new carrier design could provide important benefits for intracellular peptide drug delivery.


Asunto(s)
Micelas , Péptidos/química , Polímeros/química , Catepsina B/química , Línea Celular Tumoral , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Humanos , Concentración de Iones de Hidrógeno , Metacrilatos/química , Polietilenglicoles/química
19.
Fish Shellfish Immunol ; 60: 447-457, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27847342

RESUMEN

Cathepsin B (CTSB), a member of lysosomal cysteine protease, is involved in multiple levels of physiological and biological processes, and also plays crucial roles in host immune defense against pathogen infection in vertebrates. However, the function of CTSB within the innate immune system of invertebrates, particularly in marine echinoderms, has been poorly documented. In this study, the immune function of CTSB in Apostichopus japonicus (designated as AjCTSB), a commercially important and disease vulnerable aquaculture specie, was investigated by integrated molecular and protein approaches. A 2153 bp cDNA representing the full-length of AjCTSB was cloned via overlapping ESTs and RACE fragments. AjCTSB contained an open reading frame of 999 bp encoding a secreted protein of 332 amino acid residues with a predicted molecular mass of 36.8 kDa. The deduced amino acid of AjCTSB shared a typical activity center containing three conserved amino acid residues (Cys108, His277 and Asn297). Phylogenetic tree analysis also supported that AjCTSB was a new member of CTSB family with clustering firstly with invertebrate CTSBs. Quantitative real time PCR analysis revealed that AjCTSB was ubiquitously expressed in all examined tissues with the highest levels in intestine. The Vibrio splendidus challenged sea cucumber and LPS-exposed coelomocytes could both significantly boost the expression of AjCTSB. Moreover, the purified recombinant AjCTSB exhibited dose-dependent CTSB activities at the concentration ranged from 0 to 0.24 µg µL-1. Further functional analysis indicated that coelomocytes apoptosis was significantly inhibited by 0.16-fold in vivo and the apoptosis execution Ajcaspase 3 was extremely reduced in Apostichopus japonicus coelomocytes treated with specific AjCTSB siRNA. Collectively, all these results suggested that AjCTSB was an important immune factor and might be served as apoptosis enhancers in pathogen challenged sea cucumber.


Asunto(s)
Catepsina B/genética , Catepsina B/metabolismo , Regulación de la Expresión Génica , Inmunidad Innata , Stichopus/genética , Stichopus/inmunología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Catepsina B/química , Catepsina B/inmunología , Clonación Molecular , ADN Complementario/genética , ADN Complementario/metabolismo , Lipopolisacáridos/farmacología , Filogenia , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Alineación de Secuencia , Stichopus/microbiología , Vibrio/fisiología
20.
Biometals ; 30(2): 313-320, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28283781

RESUMEN

Gold compounds form a new class of promising metal-based drugs with a number of potential therapeutic applications, particularly in the fields of anticancer and antimicrobial treatments. Previous research revealed that a group of structurally diverse gold compounds cause conspicuous inhibition of the protease activities of the human proteasome. Given the pharmacological importance of protease inhibition, the present study further explored whether these gold compounds might inhibit a few other proteases that are accepted druggable targets for disease treatment. In particular, four distinct cysteine proteases were considered here: cathepsin B and L that play a primary role in tumor-cell invasion and metastasis; rhodesain, the major cathepsin L-like cysteine protease of Trypanosoma brucei rhodesiense and CPB2.8ΔCTE, a Leishmania mexicana mature cysteine protease. Based on the encouraging results obtained for some of the tested gold compounds on the two parasitic cysteine proteases, especially against CPB2.8ΔCTE, with IC50s in the micromolar range, we next evaluated whether those gold compounds might contrast effectively the growth of the respective protozoa and indeed important antiprotozoal properties were disclosed; on the other hand a certain lack of selectivity was highlighted. Also, no direct or clear correlation could be established between the in vitro antiprotozoal properties and the level of protease inhibition. The implications of these results are discussed in relation to possible pharmaceutical applications.


Asunto(s)
Antiprotozoarios/farmacología , Cisteína Endopeptidasas/química , Inhibidores de Cisteína Proteinasa/farmacología , Compuestos Orgánicos de Oro/farmacología , Proteínas Protozoarias/antagonistas & inhibidores , Animales , Antiprotozoarios/síntesis química , Catepsina B/antagonistas & inhibidores , Catepsina B/química , Catepsina L/antagonistas & inhibidores , Catepsina L/química , Línea Celular , Inhibidores de Cisteína Proteinasa/síntesis química , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Humanos , Concentración 50 Inhibidora , Leishmania infantum/efectos de los fármacos , Leishmania infantum/enzimología , Leishmania infantum/crecimiento & desarrollo , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/enzimología , Ratones , Compuestos Orgánicos de Oro/síntesis química , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/enzimología , Plasmodium falciparum/crecimiento & desarrollo , Complejo de la Endopetidasa Proteasomal/química , Proteínas Protozoarias/química , Proteínas Recombinantes/química , Trypanosoma brucei brucei/efectos de los fármacos , Trypanosoma brucei brucei/enzimología , Trypanosoma brucei brucei/crecimiento & desarrollo , Trypanosoma brucei rhodesiense/efectos de los fármacos , Trypanosoma brucei rhodesiense/enzimología , Trypanosoma brucei rhodesiense/crecimiento & desarrollo , Trypanosoma cruzi/efectos de los fármacos , Trypanosoma cruzi/enzimología , Trypanosoma cruzi/crecimiento & desarrollo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA