Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Biol Chem ; 405(4): 283-296, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-37889671

RESUMEN

Proteolytic activity in the tumour microenvironment is an important factor in cancer development since it can also affect intracellular signalling pathways via positive feedback loops that result in either increased tumour growth or resistance to anticancer mechanisms. In this study, we demonstrated extracellular cathepsin L-mediated cleavage of epidermal growth factor receptor (EGFR) and identified the cleavage site in the extracellular domain after R224. To further evaluate the relevance of this cleavage, we cloned and expressed a truncated version of EGFR, starting at G225, in HeLa cells. We confirmed the constitutive activation of the truncated protein in the absence of ligand binding and determined possible changes in intracellular signalling. Furthermore, we determined the effect of truncated EGFR protein expression on HeLa cell viability and response to the EGFR inhibitors, tyrosine kinase inhibitor (TKI) erlotinib and monoclonal antibody (mAb) cetuximab. Our data reveal the nuclear localization and phosphorylation of EGFR and signal trancducer and activator of transcription 3 (STAT3) in cells that express the truncated EGFR protein and suggest that these phenomena cause resistance to EGFR inhibitors.


Asunto(s)
Neoplasias Pulmonares , Humanos , Catepsina L/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos , Receptores ErbB/metabolismo , Células HeLa , Neoplasias Pulmonares/patología , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal , Microambiente Tumoral
2.
Trends Biochem Sci ; 43(10): 829-844, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30097385

RESUMEN

Proteases are considered of major importance in biomedical research because of their crucial roles in health and disease. Their ability to hydrolyze their protein and peptide substrates at single or multiple sites, depending on their specificity, makes them unique among the enzymes. Understanding protease specificity is therefore crucial to understand their biology as well as to develop tools and drugs. Recent advancements in the fields of proteomics and chemical biology have improved our understanding of protease biology through extensive specificity profiling and identification of physiological protease substrates. There are growing efforts to transfer this knowledge into clinical modalities, but their success is often limited because of overlapping protease features, protease redundancy, and chemical tools lacking specificity. Herein, we discuss the current trends and challenges in protease research and how to exploit the growing information on protease specificities for understanding protease biology, as well as for development of selective substrates, cleavable linkers, and activity-based probes and for biomarker discovery.


Asunto(s)
Péptido Hidrolasas/metabolismo , Biomarcadores/metabolismo , Conjuntos de Datos como Asunto , Humanos , Proteómica , Especificidad por Sustrato
3.
Small ; 18(12): e2107143, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35064638

RESUMEN

Ultrasound imaging with contrast agents, especially with lipid-shelled microbubbles, has become a vital tool in clinical diagnostics. Efforts to adapt these agents for molecular imaging have typically focused on targeted binding. More recently, crosslinking the lipid shell to alter its mechanical properties, followed by decrosslinking upon exposure to a stimulus, has been shown as a promising approach for imaging soluble molecular targets. Nevertheless, a systematic study of the influence of crosslinker concentration and structure on the mechanical properties of microbubbles has not been undertaken. An improved understanding of the role of these parameters is necessary to more effectively design contrast agents that detect proteases, an informative class of soluble disease markers. Here, the influence of crosslinker parameters on the acoustic properties of microbubbles, developing a model of crosslinker network formation on microbubble shells that explains the experimental observations, are studied. By incorporating cleavable elements that respond to UV light or proteolysis, kinetically resolved acoustic detection of these stimuli and the relevance of crosslinker design are demonstrated. The framework established in this study can be readily adapted to other protease-cleavable units and provides a basis for the future development of responsive ultrasound contrast agents for molecular imaging of proteolytic activity.


Asunto(s)
Medios de Contraste , Microburbujas , Acústica , Medios de Contraste/química , Lípidos/química , Ultrasonografía
4.
Proc Natl Acad Sci U S A ; 116(4): 1289-1298, 2019 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-30622175

RESUMEN

Macropinocytosis and phagocytosis are evolutionarily conserved forms of bulk endocytosis used by cells to ingest large volumes of fluid and solid particles, respectively. Both processes are regulated by Ras signaling, which is precisely controlled by mechanisms involving Ras GTPase activating proteins (RasGAPs) responsible for terminating Ras activity on early endosomes. While regulation of Ras signaling during large-scale endocytosis in WT Dictyostelium has been, for the most part, attributed to the Dictyostelium ortholog of human RasGAP NF1, in commonly used axenic laboratory strains, this gene is mutated and inactive. Moreover, none of the RasGAPs characterized so far have been implicated in the regulation of Ras signaling in large-scale endocytosis in axenic strains. In this study, we establish, using biochemical approaches and complementation assays in live cells, that Dictyostelium IQGAP-related protein IqgC interacts with active RasG and exhibits RasGAP activity toward this GTPase. Analyses of iqgC- and IqgC-overexpressing cells further revealed participation of this GAP in the regulation of both types of large-scale endocytosis and in cytokinesis. Moreover, given the localization of IqgC to phagosomes and, most prominently, to macropinosomes, we propose IqgC acting as a RasG-specific GAP in large-scale endocytosis. The data presented here functionally distinguish IqgC from other members of the Dictyostelium IQGAP family and call for repositioning of this genuine RasGAP outside of the IQGAP group.


Asunto(s)
Dictyostelium/metabolismo , Endocitosis/fisiología , Proteínas Protozoarias/metabolismo , Proteínas Activadoras de ras GTPasa/metabolismo , Secuencia de Aminoácidos , Citocinesis/fisiología , Humanos , Fagocitosis/fisiología , Fagosomas/metabolismo , Pinocitosis/fisiología , Alineación de Secuencia , Transducción de Señal/fisiología , Proteínas ras/metabolismo
5.
EMBO J ; 36(16): 2455-2465, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28733325

RESUMEN

Determination of protease specificity is of crucial importance for understanding protease function. We have developed the first gel-based label-free proteomic approach (DIPPS-direct in-gel profiling of protease specificity) that enables quick and reliable determination of protease cleavage specificities under large variety of experimental conditions. The methodology is based on in-gel digestion of the gel-separated proteome with the studied protease, enrichment of cleaved peptides by gel extraction, and subsequent mass spectrometry analysis combined with a length-limited unspecific database search. We applied the methodology to profile ten proteases ranging from highly specific (trypsin, endoproteinase GluC, caspase-7, and legumain) to broadly specific (matrix-metalloproteinase-3, thermolysin, and cathepsins K, L, S, and V). Using DIPPS, we were able to perform specificity profiling of thermolysin at its optimal temperature of 75°C, which confirmed the applicability of the method to extreme experimental conditions. Moreover, DIPPS enabled the first global specificity profiling of legumain at pH as low as 4.0, which revealed a pH-dependent change in the specificity of this protease, further supporting its broad applicability.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Proteómica/métodos , Electroforesis , Concentración de Iones de Hidrógeno , Espectrometría de Masas , Especificidad por Sustrato , Temperatura
6.
Int J Mol Sci ; 22(5)2021 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-33802262

RESUMEN

In recent decades, dysregulation of proteases and atypical proteolysis have become increasingly recognized as important hallmarks of cancer, driving community-wide efforts to explore the proteolytic landscape of oncologic disease. With more than 100 proteases currently associated with different aspects of cancer development and progression, there is a clear impetus to harness their potential in the context of oncology. Advances in the protease field have yielded technologies enabling sensitive protease detection in various settings, paving the way towards diagnostic profiling of disease-related protease activity patterns. Methods including activity-based probes and substrates, antibodies, and various nanosystems that generate reporter signals, i.e., for PET or MRI, after interaction with the target protease have shown potential for clinical translation. Nevertheless, these technologies are costly, not easily multiplexed, and require advanced imaging technologies. While the current clinical applications of protease-responsive technologies in oncologic settings are still limited, emerging technologies and protease sensors are poised to enable comprehensive exploration of the tumor proteolytic landscape as a diagnostic and therapeutic frontier. This review aims to give an overview of the most relevant classes of proteases as indicators for tumor diagnosis, current approaches to detect and monitor their activity in vivo, and associated therapeutic applications.


Asunto(s)
Neoplasias/diagnóstico , Neoplasias/metabolismo , Animales , Humanos , Péptido Hidrolasas/metabolismo , Proteolisis
7.
J Am Chem Soc ; 142(39): 16704-16715, 2020 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-32870676

RESUMEN

The subset of the proteome that contains enzymes in their catalytically active form can be interrogated by using probes targeted toward individual specific enzymes. A subset of such enzymes are proteases that are frequently studied with activity-based probes, small inhibitors equipped with a detectable tag, commonly a fluorophore. Due to the spectral overlap of these commonly used fluorophores, multiplex analysis becomes limited. To overcome this, we developed a series of protease-selective lanthanide-labeled probes compatible with mass cytometry giving us the ability to monitor the activity of multiple proteases in parallel. Using these probes, we were able to identify the distribution of four proteases with different active site geometries in three cell lines and peripheral blood mononuclear cells. This provides a framework for the use of mass cytometry for multiplexed enzyme activity detection.


Asunto(s)
Complejos de Coordinación/química , Elementos de la Serie de los Lantanoides/química , Sondas Moleculares/química , Péptido Hidrolasas/análisis , Línea Celular , Complejos de Coordinación/síntesis química , Humanos , Sondas Moleculares/síntesis química , Estructura Molecular , Péptido Hidrolasas/metabolismo
8.
Int J Mol Sci ; 21(15)2020 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-32727087

RESUMEN

(1) Background: Lipopolysaccharide (LPS)-induced systemic inflammation is associated with septic acute kidney injury (AKI). We investigated the time-dependent miRNA expression changes in the kidney caused by LPS. (2) Methods: Male outbred NMRI mice were injected with LPS and sacrificed at 1.5 and 6 h (40 mg/kg i.p., early phase, EP) or at 24 and 48 h (10 mg/kg i.p., late phase, LP). The miRNA profile was established using miRCURY LNA™ microarray and confirmed with qPCR. Total renal proteome was analyzed by LC-MS/MS (ProteomeXchange: PXD014664). (3) Results: Septic AKI was confirmed by increases in plasma urea concentration and in renal TNF-α and IL-6 mRNA expression. Most miRNAs were altered at 6 and 24 h and declined by 48 h. In EP miR-762 was newly identified and validated and was the most elevated miRNA. The predicted target of miR-762, Ras related GTPase 1B (Sar1b) was downregulated. In LP miR-21a-5p was the most influenced miRNA followed by miR-451a, miR-144-3p, and miR-146a-5p. Among the potential protein targets of the most influenced miRNAs, only aquaporin-1, a target of miR-144-3p was downregulated at 24 h. (4) Conclusion: Besides already known miRNAs, septic AKI upregulated miR-762, which may regulate GTP signaling, and miR-144-3p and downregulated its target, aquaporin-1.


Asunto(s)
Lesión Renal Aguda/metabolismo , Regulación de la Expresión Génica , MicroARNs/biosíntesis , Sepsis/metabolismo , Transcriptoma , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/patología , Animales , Lipopolisacáridos/toxicidad , Masculino , Ratones , Sepsis/inducido químicamente , Sepsis/patología
9.
J Cell Biochem ; 120(6): 10662-10669, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30652348

RESUMEN

Earlier studies showed that the oxidant menadione (MD) induces apoptosis in certain cells and also has anticancer effects. Most of these studies emphasized the role of the mitochondria in this process. However, the engagement of other organelles is less known. Particularly, the role of lysosomes and their proteolytic system, which participates in apoptotic cell death, is still unclear. The aim of this study was to investigate the role of lysosomal cathepsins on molecular signaling in MD-induced apoptosis in U937 cells. MD treatment induced translocation of cysteine cathepsins B, C, and S, and aspartic cathepsin D. Once in the cytosol, some cathepsins cleaved the proapoptotic molecule, Bid, in a process that was completely prevented by E64d, a general inhibitor of cysteine cathepsins, and partially prevented by the pancaspase inhibitor, z-VAD-fmk. Upon loss of the mitochondrial membrane potential, apoptosome activation led to caspase-9 processing, activation of caspase-3-like caspases, and poly (ADP-ribose) polymerase cleavage. Notably, the endogenous protein inhibitor, stefin B, was degraded by cathepsin D and caspases. This process was prevented by z-VAD-fmk, and partially by pepstatin A-penetratin. These findings suggest that the cleaved Bid protein acts as an amplifier of apoptotic signaling through mitochondria, thus enhancing the activity of cysteine cathepsins following stefin B degradation.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/genética , Cistatina B/genética , Regulación Neoplásica de la Expresión Génica , Lisosomas/efectos de los fármacos , Vitamina K 3/farmacología , Clorometilcetonas de Aminoácidos/farmacología , Apoptosis/genética , Apoptosomas/efectos de los fármacos , Apoptosomas/metabolismo , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Caspasa 3/genética , Caspasa 3/metabolismo , Caspasa 9/genética , Caspasa 9/metabolismo , Catepsina B/antagonistas & inhibidores , Catepsina B/genética , Catepsina B/metabolismo , Catepsina C/antagonistas & inhibidores , Catepsina C/genética , Catepsina C/metabolismo , Catepsina D/antagonistas & inhibidores , Catepsina D/genética , Catepsina D/metabolismo , Catepsinas/antagonistas & inhibidores , Catepsinas/genética , Catepsinas/metabolismo , Cistatina B/metabolismo , Humanos , Leucina/análogos & derivados , Leucina/farmacología , Lisosomas/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Pepstatinas/farmacología , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Inhibidores de Proteasas/farmacología , Proteolisis/efectos de los fármacos , Transducción de Señal , Células U937
10.
Int J Mol Sci ; 21(1)2019 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-31892161

RESUMEN

(1) Background: Sepsis-induced acute kidney injury (AKI) is the most common form of acute kidney injury (AKI). We studied the temporal profile of the sepsis-induced renal proteome changes. (2) Methods: Male mice were injected intraperitoneally with bacterial lipopolysaccharide (LPS) or saline (control). Renal proteome was studied by LC-MS/MS (ProteomeXchange: PXD014664) at the early phase (EP, 1.5 and 6 h after 40 mg/kg LPS) and the late phase (LP, 24 and 48 h after 10 mg/kg LPS) of LPS-induced AKI. Renal mRNA expression of acute phase proteins (APP) was assessed by qPCR. (3) Results: Renal proteome change was milder in EP vs. LP. APPs dominated the proteome in LP (proteins upregulated at least 4-fold (APPs/all): EP, 1.5 h: 0/10, 6 h: 1/10; LP, 24 h: 22/47, 48 h: 17/44). Lipocalin-2, complement C3, fibrinogen, haptoglobin and hemopexin were the most upregulated APPs. Renal mRNA expression preceded the APP changes with peak effects at 24 h, and indicated renal production of the majority of APPs. (4) Conclusions: Gene expression analysis revealed local production of APPs that commenced a few hours post injection and peaked at 24 h. This is the first demonstration of a massive, complex and coordinated acute phase response of the kidney involving several proteins not identified previously.


Asunto(s)
Lesión Renal Aguda/patología , Reacción de Fase Aguda/patología , Riñón/metabolismo , Riñón/patología , Proteoma/metabolismo , Sepsis/metabolismo , Sepsis/patología , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/metabolismo , Proteínas de Fase Aguda/metabolismo , Reacción de Fase Aguda/inducido químicamente , Reacción de Fase Aguda/metabolismo , Animales , Complemento C3/metabolismo , Modelos Animales de Enfermedad , Interleucina-6/metabolismo , Riñón/efectos de los fármacos , Lipopolisacáridos/farmacología , Masculino , Ratones , Sepsis/inducido químicamente , Factor de Necrosis Tumoral alfa/metabolismo
11.
Mol Cell Proteomics ; 14(8): 2213-28, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26081835

RESUMEN

Extracellular cysteine cathepsins are known to drive cancer progression, but besides degradation of extracellular matrix proteins little is known about their physiological substrates and thus the molecular mechanisms they deploy. One of the major mechanisms used by other extracellular proteases to facilitate cancer progression is proteolytic release of the extracellular domains of transmembrane proteins or ectodomain shedding. Here we show using a mass spectrometry-based approach that cathepsins L and S act as sheddases and cleave extracellular domains of CAM adhesion proteins and transmembrane receptors from the surface of cancer cells. In cathepsin S-deficient mouse pancreatic cancers, processing of these cathepsin substrates is highly reduced, pointing to an essential role of cathepsins in extracellular shedding. In addition to influencing cell migration and invasion, shedding of surface proteins by extracellular cathepsins impacts intracellular signaling as demonstrated for regulation of Ras GTPase activity, thereby providing a putative mechanistic link between extracellular cathepsin activity and cancer progression. The MS data is available via ProteomeXchange with identifier PXD002192.


Asunto(s)
Catepsina B/metabolismo , Catepsina L/metabolismo , Catepsinas/metabolismo , Membrana Celular/metabolismo , Neoplasias/metabolismo , Proteómica/métodos , Animales , Carcinogénesis/metabolismo , Carcinogénesis/patología , Línea Celular Tumoral , Movimiento Celular , Humanos , Macrófagos/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Ratones , Invasividad Neoplásica , Procesamiento Proteico-Postraduccional , Especificidad por Sustrato , Proteínas ras/metabolismo
12.
Proteomics ; 15(14): 2479-90, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25626674

RESUMEN

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).


Asunto(s)
Catepsina K/metabolismo , Catepsina L/metabolismo , Catepsinas/metabolismo , Secuencia de Aminoácidos , Catepsina K/química , Catepsina L/química , Catepsinas/química , Línea Celular Tumoral , Cromatografía Liquida/métodos , Humanos , Péptidos/química , Péptidos/metabolismo , Proteómica/métodos , Especificidad por Sustrato , Espectrometría de Masas en Tándem/métodos
13.
Angew Chem Int Ed Engl ; 53(29): 7669-73, 2014 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-24888522

RESUMEN

The synthesis and evaluation of two cathepsin S-specific probes is described. For long-term retention of the probe at the target site and a high signal-to-noise ratio, we introduced a lipidation approach via the simple attachment of palmitoic acid to the reporter. After cathepsin S-specific cleavage in cultured cells and in a grafted tumor mouse model, fluorescence increased owing to dequenching and we observed an intracellular accumulation of the fluorescence in the target tissue. The lipidated probe provided a prolonged and strongly fluorescent signal in tumors when compared to the very similar non-lipidated probe, demonstrating that non-invasive tumor identification is feasable. The homing principle by probe lipidation might also work for selective administration of cytotoxic compounds to specifically reduce tumor mass.


Asunto(s)
Catepsinas/metabolismo , Metabolismo de los Lípidos , Neoplasias Experimentales/patología , Animales , Ratones , Neoplasias Experimentales/enzimología , Especificidad por Sustrato
14.
Biol Chem ; 394(6): 783-90, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23362198

RESUMEN

Human stefins and cystatins are physiologically important cysteine proteinase inhibitors, acting as a first line of defense against undesirable proteolysis. Mutations in the cystatin B gene cause a rare form of epilepsy EPM1. Its two missense mutants, G50E and Q71P, lack the inhibitory activity and are partially unfolded, which leads to changes in their aggregation behavior, both in vitro and in the cell. SDS-PAGE and MALDI-TOF mass spectrometry were used to follow the hydrolysis of human stefin B wild type, G50E and Q71P, by cathepsins B and S in vitro. Cathepsin S was found to degrade both mutants, with Q71P being degraded faster. This correlates with the openness of the protein structure, Q71P having more exposed hydrophobic surfaces. Cathepsin B acted more selectively, degrading G50E into smaller fragments, while still leaving a portion of the full-length protein intact. Q71P was cleaved only at the exposed N-terminal end. The co-localization of stefin B wild type and EPM1 mutants with cathepsins showed that cathepsins accumulate around the aggregates formed by the EPM1 mutants. We hypothesize that the aggregation of both full-length mutants prevents the cathepsin molecule from accessing the substrate protein's core, whereas the cleaved fragments would be expected to aggregate stronger.


Asunto(s)
Catepsina B/metabolismo , Catepsinas/metabolismo , Cistatina B/química , Cistatina B/metabolismo , Proteínas Mutantes/metabolismo , Desplegamiento Proteico , Síndrome de Unverricht-Lundborg/metabolismo , Catepsinas/química , Electroforesis en Gel de Poliacrilamida , Técnica del Anticuerpo Fluorescente , Humanos , Proteínas Mutantes/química , Estabilidad Proteica , Estructura Cuaternaria de Proteína , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
15.
Cancer Metab ; 11(1): 12, 2023 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-37559137

RESUMEN

The use of bacteria in cancer immunotherapy has the potential to bypass many shortcomings of conventional treatments. The ability of anaerobic bacteria to preferentially accumulate and replicate in hypoxic regions of solid tumors, as a consequence of bacterial metabolic needs, is particularly advantageous and key to boosting their immunostimulatory therapeutic actions in situ. While several of these bacterial traits are well-studied, little is known about their competition for nutrients and its effect on cancer cells which could serve as another potent and innate antineoplastic action. Here, we explored the consequences of the iron-scavenging abilities of a particular species of bacteria, Magnetospirillum magneticum, which has been studied as a potential new class of bacteria for magnetically targeted bacterial cancer therapy. We investigated their influence in hypoxic regions of solid tumors by studying the consequential metabolic effects exerted on cancer cells. To do so, we established an in vitro co-culture system consisting of the bacterial strain AMB-1 incubated under hypoxic conditions with human breast cancer cells MDA-MB-231. We first quantified the number of viable cells after incubation with magnetotactic bacteria demonstrating a lower rate of cellular proliferation that correlated with increasing bacteria-to-cancer cells ratio. Further experiments showed increasing populations of apoptotic cells when cancer cells were incubated with AMB-1 over a period of 24 h. Analysis of the metabolic effects induced by bacteria suggest an increase in the activation of executioner caspases as well as changes in levels of apoptosis-related proteins. Finally, the level of several human apoptosis-related proteins was investigated, confirming a bacteria-dependent triggering of apoptotic pathways in breast cancer cells. Overall, our findings support that magnetotactic bacteria could act as self-replicating iron-chelating agents and indicate that they interfere with proliferation and lead to increased apoptosis of cancer cells. This bacterial feature could serve as an additional antineoplastic mechanism to reinforce current bacterial cancer therapies.

16.
Cell Chem Biol ; 30(2): 159-174.e8, 2023 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-36696904

RESUMEN

Cathepsin K (CatK) is a lysosomal cysteine protease whose highest expression is found in osteoclasts, which are the cells responsible for bone resorption. Investigations of the functions and physiological relevance of CatK have often relied on antibody-related techniques, which makes studying its activity patterns a challenging task. Hence, we developed a set of chemical tools for the investigation of CatK activity. We show that our probe is a valuable tool for monitoring the proteolytic activation of CatK during osteoclast formation. Moreover, we demonstrate that our inhibitor of CatK impedes osteoclastogenesis and bone resorption and that CatK is stored in its active form in osteoclasts within their lysosomal compartment and mainly in the ruffled borders of osteoclasts. Given that our probe recognizes active CatK within living cells without exhibiting any observed cytotoxicity in the several models tested, we expect that it would be well suited to theranostic applications in CatK-related diseases.


Asunto(s)
Resorción Ósea , Osteoclastos , Humanos , Osteoclastos/metabolismo , Osteogénesis , Catepsina K/metabolismo , Resorción Ósea/metabolismo
17.
Commun Biol ; 6(1): 450, 2023 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-37095140

RESUMEN

Addressing the elusive specificity of cysteine cathepsins, which in contrast to caspases and trypsin-like proteases lack strict specificity determining P1 pocket, calls for innovative approaches. Proteomic analysis of cell lysates with human cathepsins K, V, B, L, S, and F identified 30,000 cleavage sites, which we analyzed by software platform SAPS-ESI (Statistical Approach to Peptidyl Substrate-Enzyme Specific Interactions). SAPS-ESI is used to generate clusters and training sets for support vector machine learning. Cleavage site predictions on the SARS-CoV-2 S protein, confirmed experimentally, expose the most probable first cut under physiological conditions and suggested furin-like behavior of cathepsins. Crystal structure analysis of representative peptides in complex with cathepsin V reveals rigid and flexible sites consistent with analysis of proteomics data by SAPS-ESI that correspond to positions with heterogeneous and homogeneous distribution of residues. Thereby support for design of selective cleavable linkers of drug conjugates and drug discovery studies is provided.


Asunto(s)
COVID-19 , Cisteína , Humanos , Proteómica , SARS-CoV-2
18.
Methods Mol Biol ; 2259: 269-294, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33687722

RESUMEN

In living cells, most proteins are organized in stable or transient functional assemblies, protein complexes, which control a multitude of vital cellular processes such as cell cycle progression, metabolism, and signal transduction. Over several decades, specific protein complexes have been analyzed by structural biology methods, initially X-ray crystallography and more recently single particle cryoEM. In parallel, mass spectrometry (MS)-based methods including in vitro affinity-purification coupled to MS or in vivo protein proximity-dependent labeling methods have proven particularly effective to detect complexes, thus nominating new assemblies for structural analysis. Those approaches, however, are either of limited in throughput or require specifically engineered protein systems.In this chapter, we present protocols for a workflow that supports the parallel analysis of multiple complexes from the same biological sample with respect to abundance, subunit composition, and stoichiometry. It consists of the separation of native complexes by size-exclusion chromatography (SEC) and the subsequent mass spectrometric analysis of the proteins in consecutive SEC fractions. In particular, we describe (1) optimized conditions to achieve native protein complex separation by SEC, (2) the preparation of the SEC fractions for MS analysis, (3) the acquisition of the MS data at high throughput via SWATH/DIA (data-independent analysis) mass spectrometry and short chromatographic gradients, and (4) a set of bioinformatic tools for the targeted analysis of protein complexes. Altogether, the parallel measurement of a high number of complexes from a single biological sample results in unprecedented system-level insights into the remodeling of cellular protein complexes in response to perturbations of a broad range of cellular systems.


Asunto(s)
Cromatografía en Gel/métodos , Espectrometría de Masas/métodos , Proteínas/análisis , Proteómica/métodos , Cromatografía Líquida de Alta Presión/métodos , Humanos , Células Jurkat , Ultracentrifugación/métodos , Flujo de Trabajo
19.
Nat Commun ; 12(1): 1693, 2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33727531

RESUMEN

Proteases are among the largest protein families and critical regulators of biochemical processes like apoptosis and blood coagulation. Knowledge of proteases has been expanded by the development of proteomic approaches, however, technology for multiplexed screening of proteases within native environments is currently lacking behind. Here we introduce a simple method to profile protease activity based on isolation of protease products from native lysates using a 96FASP filter, their analysis in a mass spectrometer and a custom data analysis pipeline. The method is significantly faster, cheaper, technically less demanding, easy to multiplex and produces accurate protease fingerprints. Using the blood cascade proteases as a case study, we obtain protease substrate profiles that can be used to map specificity, cleavage entropy and allosteric effects and to design protease probes. The data further show that protease substrate predictions enable the selection of potential physiological substrates for targeted validation in biochemical assays.


Asunto(s)
Entropía , Ensayos Analíticos de Alto Rendimiento , Péptido Hidrolasas/sangre , Péptido Hidrolasas/metabolismo , Regulación Alostérica , Secuencia de Aminoácidos , Coagulación Sanguínea , Fluorescencia , Células HEK293 , Humanos , Metaloproteinasas de la Matriz/metabolismo , Péptidos/metabolismo , Especificidad por Sustrato , Tromboplastina/metabolismo
20.
Expert Opin Ther Targets ; 24(6): 573-588, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32228244

RESUMEN

Introduction: Cysteine cathepsins are involved in the development and progression of numerous inflammation-associated diseases such as cancer, arthritis, bone and immune disorders. Consequently, there is a drive to progress research efforts focused on cathepsin use in diagnostics and as therapeutic targets in disease.Areas covered: This review discusses the potential of cysteine cathepsins as therapeutic targets in inflammation-associated diseases and recent advances in preclinical and clinical research. We describe direct targeting of cathepsins for treatment purposes and their indirect use in diagnostics.Expert opinion: The targeting of cysteine cathepsins has not translated into the clinic; this failure is attributed to off- and on-target side effects and/or the lack of companion biomarkers. This field now embraces developments in diagnostic imaging, the activation of prodrugs and antibody-drug conjugates for targeted drug delivery. The future lies in improved molecular tools and therapeutic concepts that will find a wide spectrum of uses in diagnostic and therapeutic applications.


Asunto(s)
Catepsinas/metabolismo , Inflamación/tratamiento farmacológico , Terapia Molecular Dirigida , Animales , Cisteína/metabolismo , Diagnóstico por Imagen/métodos , Sistemas de Liberación de Medicamentos , Humanos , Inmunoconjugados/administración & dosificación , Inmunoconjugados/farmacología , Inflamación/diagnóstico , Inflamación/patología , Profármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA