Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Crit Rev Clin Lab Sci ; 60(4): 300-320, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36846924

RESUMEN

Collectively known as psoriatic disease, psoriasis and psoriatic arthritis (PsA) are immune-mediated inflammatory diseases in which patients present with cutaneous and musculoskeletal inflammation. Affecting roughly 2-3% of the world's total population, there remains unmet therapeutic needs in both psoriasis and PsA despite the availability of current immunomodulatory treatments. As a result, patients with psoriatic disease often experience reduced quality of life. Recently, a class of small molecules, commonly investigated as anti-cancer agents, called histone deacetylase (HDAC) inhibitors, have been proposed as a new promising anti-inflammatory treatment for immune- and inflammatory-related diseases. In inflammatory diseases, current evidence is derived from studies on diseases like rheumatoid arthritis (RA) and systematic lupus erythematosus (SLE), and while there are some reports studying psoriasis, data on PsA patients are not yet available. In this review, we provide a brief overview of psoriatic disease, psoriasis, and PsA, as well as HDACs, and discuss the rationale behind the potential use of HDAC inhibitors in the management of persistent inflammation to suggest its possible use in psoriatic disease.


Asunto(s)
Artritis Psoriásica , Psoriasis , Humanos , Artritis Psoriásica/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/uso terapéutico , Inflamación/tratamiento farmacológico , Psoriasis/tratamiento farmacológico , Calidad de Vida
2.
Pharmacol Rev ; 68(4): 1110-1142, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27677721

RESUMEN

Given that over 2% of the human genome codes for proteolytic enzymes and their inhibitors, it is not surprising that proteinases serve many physiologic-pathophysiological roles. In this context, we provide an overview of proteolytic mechanisms regulating inflammation, with a focus on cell signaling stimulated by the generation of inflammatory peptides; activation of the proteinase-activated receptor (PAR) family of G protein-coupled receptors (GPCR), with a mechanism in common with adhesion-triggered GPCRs (ADGRs); and by proteolytic ion channel regulation. These mechanisms are considered in the much wider context that proteolytic mechanisms serve, including the processing of growth factors and their receptors, the regulation of matrix-integrin signaling, and the generation and release of membrane-tethered receptor ligands. These signaling mechanisms are relevant for inflammatory, neurodegenerative, and cardiovascular diseases as well as for cancer. We propose that the inflammation-triggering proteinases and their proteolytically generated substrates represent attractive therapeutic targets and we discuss appropriate targeting strategies.

3.
Crit Rev Clin Lab Sci ; 54(7-8): 495-505, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29202618

RESUMEN

Synovial fluid (SF) is a protein-rich fluid produced into the joint cavity by cells of the synovial membrane. Due to its direct contact with articular cartilage, surfaces of the bone, and the synoviocytes of the inner membrane, it provides a promising reflection of the biochemical state of the joint under varying physiological and pathophysiological conditions. This property of SF has been exploited within numerous studies in search of unique biomarkers of joint pathologies with the ultimate goal of developing minimally invasive clinical assays to detect and/or monitor disease states. Several proteomic methodologies have been employed to mine the SF proteome. From elementary immunoassays to high-throughput analyses using mass spectrometry-based techniques, each has demonstrated distinct advantages and disadvantages in the identification and quantification of SF proteins. This review will explore the role of SF in the elucidation of the arthritis proteome and the extent to which high-throughput techniques have facilitated the discovery and validation of protein biomarkers from osteoarthritis (OA), rheumatoid arthritis (RA), psoriatic arthritis (PsA), and juvenile idiopathic arthritis (JIA) patients.


Asunto(s)
Artritis , Biomarcadores , Proteómica , Líquido Sinovial , Artritis/diagnóstico , Artritis/metabolismo , Biomarcadores/análisis , Biomarcadores/química , Humanos , Líquido Sinovial/química , Líquido Sinovial/metabolismo
4.
Clin Chem Lab Med ; 54(12): 1955-1961, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27327131

RESUMEN

BACKGROUND: Infections from microorganisms and parasites have been connected with either increased or decreased cancer risk. The objective of this study was to investigate whether infection by Echinococcus granulosus is associated with cancer risk. METHODS: We assembled a pilot retrospective cohort of patients who were diagnosed as being infected by E. granulosus in Cyprus between 1930 and 2011. Age/gender-matched non-infected family members and neighbors were selected as references. Medical history was ascertained from each study subject through in-person interview. Cox proportional hazards regression analysis was performed to assess the association of being infected by E. granulosus with cancer risk. RESULTS: Individuals with prior infection by E. granulosus (n=249) were more likely to have cancer compared to those without infection (n=753), 11.65% vs. 8.37% (p=0.0492). Survival analysis also showed that subjects with prior infection had a higher risk for developing cancer. The hazards ratio (HR) was 1.595, [95% confidence interval (CI) between 1.008 and 2.525]. The risk ratio did not change significantly (HR=1.536; 95% CI: 0.965-2.445) after adjusting for gender, year of birth, smoking status, alcohol consumption, and family history of cancer. CONCLUSIONS: Our study suggests that infection by E. granulosus may increase cancer risk. If this observation can be confirmed independently, further investigation of the mechanisms underlying the association is warranted.


Asunto(s)
Equinococosis/complicaciones , Neoplasias/complicaciones , Neoplasias/parasitología , Anciano , Animales , Estudios de Cohortes , Chipre , Equinococosis/parasitología , Femenino , Humanos , Masculino , Proyectos Piloto , Análisis de Regresión , Estudios Retrospectivos , Factores de Riesgo
5.
J Immunol ; 191(7): 3858-66, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24014879

RESUMEN

Activation of the complement system is primarily initiated by pathogen- and damage-associated molecular patterns on cellular surfaces. However, there is increasing evidence for direct activation of individual complement components by extrinsic proteinases as part of an intricate crosstalk between physiological effector systems. We hypothesized that kallikrein-related peptidases (KLKs), previously known to regulate inflammation via proteinase-activated receptors, can also play a substantial role in innate immune responses via complement. Indeed, KLKs exemplified by KLK14 were efficiently able to cleave C3, the point of convergence of the complement cascade, indicating a potential modulation of C3-mediated functions. By using in vitro fragmentation assays, mass spectrometric analysis, and cell signaling measurements, we pinpointed the generation of the C3a fragment of C3 as a product with potential biological activity released by the proteolytic action of KLK14. Using mice with various complement deficiencies, we demonstrated that the intraplantar administration of KLK14 results in C3-associated paw edema. The edema response was dependent on the presence of the receptor for C3a but was not associated with the receptor for the downstream complement effector C5a. Our findings point to C3 as one of the potential substrates of KLKs during inflammation. Given the wide distribution of the KLKs in tissues and biological fluids where complement components may also be expressed, we suggest that via C3 processing, tissue-localized KLKs can play an extrinsic complement-related role during activation of the innate immune response.


Asunto(s)
Calicreínas/metabolismo , Receptores de Complemento/metabolismo , Animales , Activación de Complemento/inmunología , Complemento C3/genética , Complemento C3/inmunología , Complemento C3/metabolismo , Complemento C3a/inmunología , Complemento C3a/metabolismo , Complemento C5/genética , Complemento C5/inmunología , Complemento C5/metabolismo , Modelos Animales de Enfermedad , Edema/genética , Edema/inmunología , Edema/metabolismo , Humanos , Calicreínas/administración & dosificación , Calicreínas/inmunología , Ratones , Ratones Noqueados , Proteolisis , Receptores de Complemento/genética
6.
Crit Rev Clin Lab Sci ; 51(3): 138-48, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24588712

RESUMEN

Infectious agents have been associated with cancer due to activation of pro-carcinogenic inflammatory processes within their host. Several reports, however, indicate that specific pathogens may be able to elicit anti-tumor immune responses that can lead to protection from tumorigenesis or cancer regression. Amongst these "beneficial" pathogens are some helminthic parasites that have already been connected with prevention of autoimmune diseases and allergies, immune conditions increasingly associated with cancer. Even though helminths have co-existed with humans and their ancestors for millions of years, investigations of their impact on human (patho)physiology are relatively new and the functions of components that can explain the helminth bi-directional influence on carcinogenesis are not well understood. This review aims to discuss evidence for the helminth-induced immune, genetic, epigenetic, proteomic, hormonal and metabolic changes that may ultimately mediate the potential pro- or anti-carcinogenic role of helminths. This overview may serve future investigations in clarifying the tumorigenic role of the most common helminthic parasites. It may also inspire the development of anti-cancer regimens and vaccines, in parallel to ongoing efforts of using helminth-based components for the prevention and/or treatment of autoimmune diseases and allergies.


Asunto(s)
Carcinogénesis/patología , Helmintiasis/patología , Helmintos/aislamiento & purificación , Neoplasias/parasitología , Animales , Interacciones Huésped-Parásitos , Humanos , Inflamación/parasitología
7.
Biol Chem ; 395(9): 1075-86, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25153388

RESUMEN

Emerging evidence indicates that serine proteases of the tissue kallikrein-related peptidases family (KLK) are implicated in tumorigenesis. We recently reported the ectopic expression of KLK4 and KLK14 in colonic cancers and their signaling to control cell proliferation. Human tissue kallikrein-related peptidase 7 (KLK7) is often dysregulated in many cancers; however, its role in colon tumorigenesis has not yet been established. In the present study, we analyzed expression of KLK7 in 15 colon cancer cell lines and in 38 human colonic tumors. In many human colon cancer cells, KLK7 mRNA was observed, which leads to KLK7 protein expression and secretion. Furthermore, KLK7 was detected in human colon adenocarcinomas, but it was absent in normal epithelia. KLK7 overexpression in HT29 colon cancer cells upon stable transfection with a KLK7 expression plasmid resulted in increased cell proliferation. Moreover, subcutaneous inoculation of transfected cells into nude mice led to increased tumor growth that was associated with increased tumor cell proliferation as reflected by a positive Ki-67 staining. Our results demonstrate the aberrant expression of KLK7 in colon cancer cells and tissues and its involvement in cell proliferation in vitro and in vivo. Thus, KLK7 may represent a potential therapeutic target for human colon tumorigenesis.


Asunto(s)
Neoplasias del Colon/genética , Neoplasias del Colon/patología , Calicreínas/genética , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Células HT29 , Humanos , Ratones , Ratones Desnudos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transfección , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Arthritis Res Ther ; 26(1): 107, 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38802975

RESUMEN

BACKGROUND: Psoriatic arthritis (PsA) is an inflammatory arthritis associated with psoriasis. PsA disease involves flares, which are associated with increased joint inflammation and tissue remodeling. There is a need for identifying biomarkers related to PsA disease activity and flares to improve the management of PsA patients and decrease flares. The tissue turnover imbalance that occurs during the inflammatory and fibro-proliferative processes during flares leads to an increased degradation and/or reorganization of the extracellular matrix (ECM), where increased proteolysis plays a key role. Hence, protease-mediated fragments of inflammatory and tissue-remodeling components could be used as markers reflecting flares in PsA patients. METHODS: A broad panel of protease-mediated biomarkers reflecting inflammation and tissue remodeling was measured in serum and synovial fluid (SF) obtained from PsA patients experiencing flares (acutely swollen joint[s], PsA-flare). In serum, biomarker levels assessed in PsA-flare patients were compared to controls and in early-diagnosed PsA patients not experiencing flares (referred to as PsA without flare). Furthermore, the biomarker levels assessed in SF from PsA-flare patients were compared to the levels in SF of osteoarthritis (OA) patients. RESULTS: In serum, levels of the PRO-C3 and C3M, reflecting formation and degradation of the interstitial matrix, were found significantly elevated in PsA-flare compared to controls and PsA without flare. The remodeling marker of the basement membrane, PRO-C4, was significantly elevated in PsA-flare compared to PsA without flare. The inflammation and immune cell activity related markers, CRPM, VICM, and CPa9-HNE were significantly elevated in PsA-flare patients compared to controls and PsA without flare. In addition, VICM (AUC = 0.71), CPa9-HNE (AUC = 0.89), CRPM (AUC = 0.76), and PRO-C3 (AUC = 0.86) showed good discriminatory performance for separating PsA-flare from PsA without flare. In SF, the macrophage activity marker, VICM, was significantly elevated whereas the type II collagen formation marker, PRO-C2, was significantly reduced in the PsA-flare compared to OA. The combination of five serum markers reflecting type III and IV collagen degradation (C3M and C4M, respectively), type III and VI collagen formation (PRO-C3 and PRO-C6, respectively), and neutrophil activity (CPa9-HNE) showed an excellent discriminatory performance (AUC = 0.98) for separating PsA-flare from PsA without flares. CONCLUSIONS: The serum biomarker panel of C3M, C4M, PRO-C3, PRO-C6, and CPa9-HNE reflecting synovitis, enthesitis, and neutrophil activity may serve as novel tool for quantitatively monitoring flares in PsA patients.


Asunto(s)
Artritis Psoriásica , Biomarcadores , Humanos , Artritis Psoriásica/sangre , Artritis Psoriásica/diagnóstico , Artritis Psoriásica/metabolismo , Biomarcadores/sangre , Masculino , Femenino , Persona de Mediana Edad , Adulto , Líquido Sinovial/metabolismo , Péptido Hidrolasas/sangre , Péptido Hidrolasas/metabolismo , Inflamación/sangre , Inflamación/metabolismo , Anciano , Péptidos/sangre
9.
Mediators Inflamm ; 2013: 713284, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23737652

RESUMEN

Acylation stimulating protein (ASP) is an adipokine derived from the immune complement system, which stimulates fat storage and is typically increased in obesity, type 2 diabetes, and cardiovascular disease. Using a diet-induced obesity (DIO) mouse model, the acute effects of ASP on energy metabolism and inflammatory processes in vivo were evaluated. We hypothesized that ASP would specifically exert proinflammatory effects. C57Bl/6 wild-type mice were put on a high-fat-high-sucrose diet for 12 weeks. Mice were then subjected to both glucose and insulin tolerance tests, each manipulation being preceded by recombinant ASP or vehicle (control) bolus injection. ASP supplementation increased whole-body glucose excursion, and this was accomplished with reduced concomitant insulin levels. However, ASP did not directly alter insulin sensitivity. ASP supplementation induced a proinflammatory phenotype, with higher levels of cytokines including IL-6 and TNF-α in plasma and in adipose tissue, liver, and skeletal muscle mRNA. Additionally, ASP increased M1 macrophage content of these tissues. ASP exerted a direct concentration-dependent role in the migration and M1 activation of cultured macrophages. Altogether, the in vivo and in vitro experiments demonstrate that ASP plays a role in both energy metabolism and inflammation, with paradoxical whole-body glucose-sensitizing yet proinflammatory effects.


Asunto(s)
Complemento C3a/farmacología , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Grasas de la Dieta/efectos adversos , Humanos , Insulina/farmacología , Resistencia a la Insulina/fisiología , Interleucina-6/sangre , Macrófagos/citología , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/sangre , Obesidad/etiología , Obesidad/metabolismo , Factor de Necrosis Tumoral alfa/sangre
10.
J Biol Chem ; 286(29): 25505-18, 2011 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-21628462

RESUMEN

Kallikrein-related peptidases (KLKs) are an emerging group of secreted serine proteases involved in several physiological and pathological processes. We used a degradomic approach to identify potential substrates of KLK12. MDA-MB-231 cells were treated either with KLK12 or vehicle control, and the proteome of the overlying medium was analyzed by mass spectrometry. CCN1 (cyr61, ctgf, nov) was among the proteins released by the KLK12-treated cells, suggesting that KLK12 might be responsible for the shedding of this protein from the cell surface. Fragmentation of CCN1 by KLK12 was further confirmed in vitro, and the main cleavage site was localized in the hinge region between the first and second half of the recombinant protein. KLK12 can target all six members of the CCN family at different proteolytic sites. Limited proteolysis of CCNs (cyr61, ctgf, nov) was also observed in the presence of other members of the KLK family, such as KLK1, KLK5, and KLK14, whereas KLK6, KLK11, and KLK13 were unable to fragment CCNs. Because KLK12 seems to have a role in angiogenesis, we investigated the relations between KLK12, CCNs, and several factors known to be involved in angiogenesis. Solid phase binding assays showed that fragmentation of CCN1 or CCN5 by KLK12 prevents VEGF(165) binding, whereas it also triggers the release of intact VEGF and BMP2 from the CCN complexes. The KLK12-mediated release of TGF-ß1 and FGF-2, either as intact or truncated forms, was found to be concentration-dependent. These findings suggest that KLK12 may indirectly regulate the bioavailability and activity of several growth factors through processing of their CCN binding partners.


Asunto(s)
Proteína 61 Rica en Cisteína/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Calicreínas/metabolismo , Calicreínas/farmacología , Factores de Transcripción/metabolismo , Proteínas CCN de Señalización Intercelular , Línea Celular Tumoral , Proteína 61 Rica en Cisteína/genética , Regulación Neoplásica de la Expresión Génica , Homeostasis/efectos de los fármacos , Humanos , Hidrólisis , Péptidos y Proteínas de Señalización Intercelular/genética , Calicreínas/genética , Pulmón/citología , Pulmón/metabolismo , Pulmón/patología , Neoplasias Pulmonares/genética , Unión Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteómica , Proteínas Represoras , Reproducibilidad de los Resultados , Factores de Transcripción/genética
11.
Biol Chem ; 393(5): 413-20, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22505523

RESUMEN

We hypothesized that kallikrein-related peptidase 14 (KLK14) is produced by colonic tumors and can promote tumorigenesis by activating proteinase-activated receptors (PARs). We found that KLK14 is expressed in human colon adenocarcinoma cells but not in adjacent cancer-free tissue; KLK14 mRNA, present in colon cancer, leads to KLK14 protein expression and secretion; and KLK14 signals viaPAR-2 in HT-29 cells to cause (1) receptor activation/internalization, (2) increases in intracellular calcium, (3) stimulation of ERK1/2/MAP kinase phosphorylation, and (4) cell proliferation. We suggest that KLK14, acting via PAR-2, represents an autocrine/paracrine regulator of colon tumorigenesis.


Asunto(s)
Neoplasias del Colon/patología , Calicreínas/metabolismo , Receptor PAR-2/metabolismo , Transducción de Señal , Neoplasias del Colon/enzimología , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Regulación Neoplásica de la Expresión Génica , Células HT29 , Humanos , Calicreínas/genética , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo
12.
Biol Chem ; 393(5): 421-7, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22505524

RESUMEN

We compared signalling pathways such as calcium transients, MAPK activation, ß-arrestin interactions and receptor internalization triggered by kallikrein-related peptidases (KLKs) 8 and 14 in human and rat proteinase-activated receptor (PAR)2-expressing human embryonic kidney (HEK) and Kirsten transformed rat kidney (KNRK) cells. Further, we analysed processing by KLK8 vs. KLK14 of synthetic human and rat PAR2-derived sequences representing the cleavage-activation domain of PAR2. Our data show that like KLK14, KLK8 can unmask a PAR2 receptor-activating sequence from a peptide precursor. However, whilst KLK8, like KLK14, can signal in rat-PAR2-expressing KNRK cells, this enzyme cannot signal via human PAR2 in HEK or KNRK cells, but rather, disarms HEK PAR1. Thus, KLK8 and KLK14 can signal differentially via the PARs to affect tissue function.


Asunto(s)
Calicreínas/metabolismo , Receptor PAR-2/metabolismo , Transducción de Señal , Animales , Células HEK293 , Humanos , Transporte de Proteínas , Ratas
13.
Am J Pathol ; 179(5): 2625-36, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21907696

RESUMEN

Serine proteinases participate in tumor growth and invasion by cleaving and activating proteinase-activated receptors (PARs). Recent studies have implicated PAR-1 and PAR-4 (activated by thrombin) and PAR-2 (activated by trypsin but not by thrombin) in human colon cancer growth. The endogenous activators of PARs in colon tumors, however, are still unknown. We hypothesize that the kallikrein-related peptidase (KLK) family member KLK14, a known tumor biomarker, is produced by colonic tumors and signals to human colon cancer cells by activating PARs. We found that i) KLK14 mRNA was present in 16 human colon cancer cell lines, ii) KLK14 protein was expressed and secreted in colon cancer cell lines, and iii) KLK14 (0.1 µmol/L) induced increases in intracellular calcium in HT29, a human colon cancer-derived cell line. KLK14-induced calcium flux was associated with internalization of KLK14-mediated activation of PAR-2. Furthermore, KLK14 induced significant extracellular signal-regulated kinases 1 and 2 (ERK1/2) phosphorylation and HT29 cell proliferation, presumably by activating PAR-2. A PAR-2 cleavage and activation-blocking antibody dramatically reduced KLK14-induced ERK1/2 signaling. Finally, ectopic expression of KLK14 in human colon adenocarcinomas and its absence in normal epithelia was demonstrated by IHC analysis. These results demonstrate, for the first time, the aberrant expression of KLK14 in colon cancer and its involvement in PAR-2 receptor signaling. Thus, KLK14 and its receptor, PAR-2, may represent therapeutic targets for colon tumorigenesis.


Asunto(s)
Adenocarcinoma/enzimología , Neoplasias del Colon/enzimología , Calicreínas/fisiología , Receptor PAR-2/metabolismo , Transducción de Señal/fisiología , Señalización del Calcio/fisiología , Células HT29 , Humanos , Calicreínas/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Regulación hacia Arriba
14.
Biomolecules ; 12(7)2022 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-35883559

RESUMEN

Kallikrein-related peptidases (KLKs) are implicated in many cancer-related processes. KLK6, one of the 15 KLK family members, is a promising biomarker for diagnosis of many cancers and has been associated with poor prognosis of colorectal cancer (CRC) patients. Herein, we evaluated the expression and cellular functions of KLK6 in colon cancer-derived cell lines and in clinical samples from CRC patients. We showed that, although many KLKs transcripts are upregulated in colon cancer-derived cell lines, KLK6, KLK10, and KLK11 are the most highly secreted proteins. KLK6 induced calcium flux in HT29 cells by activation and internalization of protease-activated receptor 2 (PAR2). Furthermore, KLK6 induced extracellular signal-regulated kinases 1 and 2 (ERK1/2) phosphorylation. KLK6 suppression in HCT-116 colon cancer cells decreased the colony formation, increased cell adhesion to extracellular matrix proteins, and reduced spheroid formation and compaction. Immunohistochemistry (IHC) analysis demonstrated ectopic expression of KLK6 in human colon adenocarcinomas but not in normal epithelia. Importantly, high levels of KLK6 protein were detected in the ascites of CRC patients with peritoneal metastasis, but not in benign ascites. These data indicate that KLK6 overexpression is associated with aggressive CRC, and may be applied to differentiate between benign and malignant ascites.


Asunto(s)
Neoplasias del Colon , Neoplasias Peritoneales , Neoplasias del Recto , Ascitis , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Humanos , Calicreínas/genética , Calicreínas/metabolismo , Fenotipo
16.
Biol Chem ; 391(4): 299-310, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20180639

RESUMEN

The exact mechanism(s) by which kallikrein-related peptidases (KLKs) function, their levels of activity and their potential endogenous targets in vivo have only recently begun to be revealed. Our group and others have shown that KLKs can have hormonal properties by signaling via proteinase-activated receptors (PARs), a family of G-protein-coupled receptors. Signals by PAR(1), PAR(2), and PAR(4) can regulate calcium release or mitogen-activated protein kinase activation and lead to platelet aggregation, vascular relaxation, cell proliferation, cytokine release, and inflammation. We have further documented the presence of active KLK6 and 10 (by activity-based ELISA or proteomics) and the presence of proteinase inhibitors, such as alpha(1)-antitrypsin, in cancer-derived fluids. We suggest that tumors and inflamed tissues can release active KLKs, which are under tight regulation by proteinase inhibitors. These enzymes can potentially control cell/tissue behavior by regulating PAR activation in specific settings and disease stages.


Asunto(s)
Calicreínas/metabolismo , Neoplasias/enzimología , Neoplasias/metabolismo , Transducción de Señal , Animales , Hormonas/metabolismo , Humanos , Inflamación/metabolismo , Neoplasias/patología , Receptores Proteinasa-Activados/metabolismo
17.
Biol Chem ; 391(4): 381-90, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20180649

RESUMEN

Kallikrein-related peptidases (KLKs) are secreted serine proteinases with trypsin or chymotrypsin-like activity. Several family members, such as KLKs 6 and 10, are potential ovarian cancer biomarkers. Recently, using a newly developed assay for active KLK6, we found that only a very small proportion of immunoreactive KLK6 in tumor-derived clinical samples (malignant ascites fluid), in cerebrospinal fluid, and in cancer cell line supernatants is enzymatically active. We therefore hypothesized that a proportion of other immunoreactive KLKs in such samples could be present, but might be partly complexed to endogenous serine proteinase inhibitors. Using a combination of immunological isolation of the enzymes, activity-based probe analysis and proteomics, we identified active KLK10 in ovarian cancer ascites and we provide preliminary data that the activity of other KLKs present in these samples can be decreased by known proteinase inhibitors (e.g., alpha2-macroglobulin, alpha1-antitrypsin). Our data suggest that the enzymatic activity of ovarian cancer-released KLKs that are detected by regular immunoassays is low in vivo and very likely regulated by proteinase inhibitors.


Asunto(s)
Ascitis/enzimología , Calicreínas/metabolismo , Neoplasias Ováricas/enzimología , Proteómica/métodos , Cromatografía en Gel , Femenino , Humanos , Calicreínas/química , Sondas Moleculares/metabolismo , Peso Molecular , Inhibidores de Proteasas/metabolismo , Tripsina/metabolismo
18.
Clin Rheumatol ; 39(8): 2355-2361, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32100195

RESUMEN

INTRODUCTION/OBJECTIVES: Intra-articular corticosteroid (IAS) injections are often used for the immediate relief of pain and inflammation in the joint of psoriatic arthritis (PsA) patients. However, studies identifying factors that predict response to the IAS injections are lacking. We aimed to assess the usefulness of serine proteinase activity measurements in PsA synovial fluid (SF) samples obtained at the time of injection in predicting clinical response. METHODS: The PsA patients with available SF samples from the knee joint were identified from the University of Toronto PsA cohort. Clinical response was defined as an absence of tenderness or swelling in the injected joint at the first post-injection visit, at either 3 or 6 months. SF proteinase activity was determined by measuring cleavage of fluorogenic tri-peptide substrates for trypsin-like (VPR-AMC and VLK-AMC) and chymotrypsin-like (AAPF-AMC) serine proteinases. Generalized estimating equation (GEE) models were used to investigate factors associated with response. RESULTS: A total of 32 patients with 60 injected joints and data available for follow-up at 3 or 6 months were included in the analysis, with 25 (41.7%) injected joints resulting in clinical response. Age, sex, active joint count, systemic medications and SF serine proteinase activity at the time of injection were included as covariates. Only treatment with biologics was significantly associated with response at 3 or 6 months in the multivariate reduced model (OR 3.02, p = 0.027). CONCLUSIONS: We could not demonstrate an association between SF serine proteinase activity and response to IAS injection. Biologic agents significantly improve the likelihood of achieving clinical response.


Asunto(s)
Corticoesteroides/administración & dosificación , Artritis Psoriásica/tratamiento farmacológico , Artritis Psoriásica/metabolismo , Serina Proteasas/metabolismo , Adulto , Artritis Psoriásica/patología , Biomarcadores/metabolismo , Monitoreo de Drogas/métodos , Femenino , Humanos , Inyecciones Intraarticulares , Articulación de la Rodilla/metabolismo , Articulación de la Rodilla/patología , Masculino , Persona de Mediana Edad , Análisis Multivariante , Estudios Prospectivos , Líquido Sinovial/metabolismo
19.
Front Immunol ; 11: 629726, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33763056

RESUMEN

Objective: Multiple proteinases are present in the synovial fluid (SF) of an arthritic joint. We aimed to identify inflammatory cell populations present in psoriatic arthritis (PsA) SF compared to osteoarthritis (OA) and rheumatoid arthritis (RA), identify their proteinase-activated receptor 2 (PAR2) signaling function and characterize potentially active SF serine proteinases that may be PAR2 activators. Methods: Flow cytometry was used to characterize SF cells from PsA, RA, OA patients; PsA SF cells were further characterized by single cell 3'-RNA-sequencing. Active serine proteinases were identified through cleavage of fluorogenic trypsin- and chymotrypsin-like substrates, activity-based probe analysis and proteomics. Fluo-4 AM was used to monitor intracellular calcium cell signaling. Cytokine expression was evaluated using a multiplex Luminex panel. Results: PsA SF cells were dominated by monocytes/macrophages, which consisted of three populations representing classical, non-classical and intermediate cells. The classical monocytes/macrophages were reduced in PsA compared to OA/RA, whilst the intermediate population was increased. PAR2 was elevated in OA vs. PsA/RA SF monocytes/macrophages, particularly in the intermediate population. PAR2 expression and signaling in primary PsA monocytes/macrophages significantly impacted the production of monocyte chemoattractant protein-1 (MCP-1). Trypsin-like serine proteinase activity was elevated in PsA and RA SF compared to OA, while chymotrypsin-like activity was elevated in RA compared to PsA. Tryptase-6 was identified as an active serine proteinase in SF that could trigger calcium signaling partially via PAR2. Conclusion: PAR2 and its activating proteinases, including tryptase-6, can be important mediators of inflammation in PsA. Components within this proteinase-receptor axis may represent novel therapeutic targets.


Asunto(s)
Artritis Psoriásica/inmunología , Señalización del Calcio/inmunología , Macrófagos/inmunología , Receptor PAR-2/inmunología , Triptasas/inmunología , Artritis Psoriásica/patología , Femenino , Humanos , Macrófagos/patología , Masculino
20.
Adv Cancer Res ; 96: 23-50, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17161675

RESUMEN

Despite impressive scientific achievements over the past few decades, cancer is still a leading cause of death. One of the major reasons is that most cancer patients are diagnosed with advanced disease. This is clearly illustrated with ovarian cancer in which the overall 5-year survival rates are only 20-30%. Conversely, when ovarian cancer is detected early (stage 1), the 5-year survival rate increases to 95%. Biomarkers, as tools for preclinical detection of cancer, have the potential to revolutionize the field of clinical diagnostics. The emerging field of clinical proteomics has found applications across a wide spectrum of cancer research. This chapter will focus on mass spectrometry as a proteomic technology implemented in three areas of cancer: diagnostics, tissue imaging, and biomarker discovery. Despite its power, it is also important to realize the preanalytical, analytical, and postanalytical limitations currently associated with this methodology. The ultimate endpoint of clinical proteomics is individualized therapy. It is essential that research groups, the industry, and physicians collaborate to conduct large prospective, multicenter clinical trials to validate and standardize this technology, for it to have real clinical impact.


Asunto(s)
Biomarcadores de Tumor/análisis , Espectrometría de Masas/métodos , Neoplasias/genética , Proteoma , Proteómica/métodos , Animales , Femenino , Humanos , Tamizaje Masivo/métodos , Neoplasias/diagnóstico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA