Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 625
Filtrar
1.
Int J Mol Sci ; 21(18)2020 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-32948029

RESUMEN

The crucial role of extracellular proteases in cancer progression is well-known, especially in relation to the promotion of cell invasion through extracellular matrix remodeling. This also occurs by the ability of extracellular proteases to induce the shedding of transmembrane proteins at the plasma membrane surface or within extracellular vesicles. This process results in the regulation of key signaling pathways by the modulation of kinases, e.g., the epidermal growth factor receptor (EGFR). Considering their regulatory roles in cancer, therapeutics targeting various extracellular proteases have been discovered. These include the metal-binding agents di-2-pyridylketone 4,4-dimethyl-3-thiosemicarbazone (Dp44mT) and di-2-pyridylketone-4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC), which increase c-MET degradation by multiple mechanisms. Both the direct and indirect inhibition of protease expression and activity can be achieved through metal ion depletion. Considering direct mechanisms, chelators can bind zinc(II) that plays a catalytic role in enzyme activity. In terms of indirect mechanisms, Dp44mT and DpC potently suppress the expression of the kallikrein-related peptidase-a prostate-specific antigen-in prostate cancer cells. The mechanism of this activity involves promotion of the degradation of the androgen receptor. Additional suppressive mechanisms of Dp44mT and DpC on matrix metalloproteases (MMPs) relate to their ability to up-regulate the metastasis suppressors N-myc downstream regulated gene-1 (NDRG1) and NDRG2, which down-regulate MMPs that are crucial for cancer cell invasion.


Asunto(s)
Antineoplásicos/uso terapéutico , Quelantes/uso terapéutico , Hierro , Proteínas de Neoplasias/fisiología , Péptido Hidrolasas/fisiología , Inhibidores de Proteasas/uso terapéutico , Zinc , Antineoplásicos/farmacología , Línea Celular Tumoral , Transformación Celular Neoplásica , Quelantes/farmacología , Progresión de la Enfermedad , Diseño de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Líquido Extracelular/enzimología , Vesículas Extracelulares/enzimología , Humanos , Ácidos Hidroxámicos/farmacología , Ácidos Hidroxámicos/uso terapéutico , Quelantes del Hierro/farmacología , Quelantes del Hierro/uso terapéutico , Calicreínas/antagonistas & inhibidores , Calicreínas/fisiología , Metaloproteinasas de la Matriz/fisiología , Terapia Molecular Dirigida , Proteínas de Neoplasias/antagonistas & inhibidores , Oxaprozina/farmacología , Oxaprozina/uso terapéutico , Fenilalanina/análogos & derivados , Fenilalanina/farmacología , Fenilalanina/uso terapéutico , Inhibidores de Proteasas/farmacología , Proteínas Quinasas/fisiología , Piridinas/farmacología , Piridinas/uso terapéutico , Tiofenos/farmacología , Tiofenos/uso terapéutico , Tiosemicarbazonas/farmacología , Tiosemicarbazonas/uso terapéutico
2.
Neoplasma ; 67(4): 889-897, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32386481

RESUMEN

Colorectal cancer (CRC) is a common aggressive carcinoma with a proverbial feature of metabolic reprogramming that is essential for cancer cell growth. Recent research corroborates the controversial function of kallikrein-related peptidase 10 (KLK10) in cancer. However, its role and underlying mechanism in CRC remains elusive. In the present study, high expression of KLK10 was detected in CRC cell lines. Knockdown of KLK10 expression by a specific siRNA inhibited cell proliferation, evoked cell apoptosis, and increased caspase-3 activity in HT29 CRC cells. Furthermore, KLK10 suppression also afforded the suppressive effects on glycolysis in CRC cells as the data showed that targeting KLK10 restrained glucose uptake, lactate production, and glycolysis-related glucose transporter 1 (Glut1) expression. Mechanism analysis corroborated that cessation of KLK10 muted the PI3K/AKT-mTOR signaling. Intriguingly, reactivating the PI3K/AKT-mTOR pathway by its agonist IGF-1 notably reversed the inhibitory effects of KLK10 cessation on CRC cell growth and glucose metabolism. More important, preconditioning with PI3K/AKT inhibitor LY294002 or mTOR inhibitor rapamycin both aggravated KLK10 knockdown-suppressed cancer cell growth and glucose metabolism. These findings suggest that KLK10 silencing may attenuate the progression of CRC by inhibiting cell growth and glycolysis via the PI3K/AKT/mTOR signaling, supporting a potential and promising target for CRC therapy.


Asunto(s)
Neoplasias Colorrectales , Calicreínas , Proteínas Proto-Oncogénicas c-akt , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Regulación de la Expresión Génica , Silenciador del Gen , Glucosa/metabolismo , Humanos , Calicreínas/fisiología , Fosfatidilinositol 3-Quinasas , Serina-Treonina Quinasas TOR
3.
Presse Med ; 48(1 Pt 1): 55-62, 2019 Jan.
Artículo en Francés | MEDLINE | ID: mdl-30416009

RESUMEN

Bradykinin mediated angioedema (BK-AE) can be associated either with C1Inhibitor deficiency (hereditary and acquired forms), either with normal C1Inh (hereditary form and drug induced AE as angiotensin converting enzyme inhibitors…). In case of high clinical suspicion of BK-AE, C1Inh exploration must be done at first: C1Inh function and antigenemy as well as C4 concentration. C1Inh deficiency is significant if the tests are below 50 % of the normal values and controlled a second time. In case of C1Inh deficiency, you have to identify hereditary from acquired forms. C1q and anti-C1Inh antibody tests are useful for acquired BK-AE. SERPING1 gene screening must be done if a hereditary angioedema is suspected, even if there is no family context (de novo mutation 15 %). If a hereditary BK-AE with normal C1Inh is suspected, F12 and PLG gene screening is suitable.


Asunto(s)
Angioedemas Hereditarios/metabolismo , Bradiquinina/metabolismo , Proteína Inhibidora del Complemento C1/análisis , Algoritmos , Angioedema/inducido químicamente , Angioedema/metabolismo , Angioedemas Hereditarios/clasificación , Inhibidores de la Enzima Convertidora de Angiotensina/efectos adversos , Niño , Comorbilidad , Proteína Inhibidora del Complemento C1/genética , Diagnóstico Precoz , Factor XII/fisiología , Femenino , Fibrinolisina/fisiología , Enfermedades Hematológicas/epidemiología , Angioedema Hereditario Tipos I y II/diagnóstico , Angioedema Hereditario Tipos I y II/metabolismo , Humanos , Calicreínas/fisiología , Lupus Eritematoso Sistémico/epidemiología , Embarazo , Complicaciones del Embarazo/sangre , Evaluación de Síntomas
4.
J Thromb Haemost ; 16(9): 1674-1685, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29920929

RESUMEN

The plasma contact system contributes to thrombosis in experimental models. Even though our standard blood coagulation tests are prolonged when plasma lacks contact factors, this enzyme system appears to have a minor (if any) role in hemostasis. In this review, we explore the clinical phenotype of C1 esterase inhibitor (C1-INH) deficiency. C1-INH is the key plasma inhibitor of the contact system enzymes, and its deficiency causes hereditary angioedema (HAE). This inflammatory disorder is characterized by recurrent aggressive attacks of tissue swelling that occur at unpredictable locations throughout the body. Bradykinin, which is considered to be a byproduct of the plasma contact system during in vitro coagulation, is the main disease mediator in HAE. Surprisingly, there is little evidence for thrombotic events in HAE patients, suggesting mechanistic uncoupling from the intrinsic pathway of coagulation. In addition, it is questionable whether a surface is responsible for contact system activation in HAE. In this review, we discuss the clinical phenotype, disease modifiers and diagnostic challenges of HAE. We subsequently describe the underlying biochemical mechanisms and contributing disease mediators. Furthermore, we review three types of HAE that are not caused by C1-INH inhibitor deficiency. Finally, we propose a central enzymatic axis that we hypothesize to be responsible for bradykinin production in health and disease.


Asunto(s)
Angioedemas Hereditarios/sangre , Coagulación Sanguínea/fisiología , Bradiquinina/fisiología , Edad de Inicio , Angioedemas Hereditarios/enzimología , Angioedemas Hereditarios/etiología , Angioedemas Hereditarios/fisiopatología , Bradiquinina/biosíntesis , Permeabilidad Capilar , Activación de Complemento , Proteína Inhibidora del Complemento C1/fisiología , Factor XIIa/fisiología , Femenino , Angioedema Hereditario Tipos I y II/sangre , Angioedema Hereditario Tipos I y II/enzimología , Angioedema Hereditario Tipos I y II/fisiopatología , Humanos , Inflamación , Calidina/metabolismo , Calicreínas/fisiología , Quininógeno de Alto Peso Molecular/metabolismo , Masculino , Modelos Biológicos , Fenotipo , Polifosfatos/metabolismo , Inhibidores de Serina Proteinasa/deficiencia , Inhibidores de Serina Proteinasa/fisiología
5.
Sci Rep ; 8(1): 6331, 2018 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-29679011

RESUMEN

Kallikrein-related peptidase 12 (KLK12) is a kallikrein family peptidase involved in angiogenesis - a complex biological process in which the sprouting, migration and stabilization of endothelial cells requires extracellular matrix remodeling. To characterize the molecular mechanisms associated with KLK12's proangiogenic activity, we evaluated its ability to hydrolyze various matrix proteins. Our results show that KLK12 efficiently cleaved the human extracellular matrix proteins fibronectin and tenascin, both of which are involved in the regulation of endothelial cell adhesion and migration. For fibronectin, the major proteolytic product generated by KLK12 was a 29 kDa fragment containing the amino-terminal domain and the first five type I fibronectin-domains, which are essential for regulating fibronectin assembly. We also demonstrated that KLK12-mediated fibronectin proteolysis antagonizes fibronectin polymerization and fibronectin fibril formation by endothelial cells, leading to an increase in cell migration. Furthermore, a polyclonal antibody raised against KLK12's proteolytic cleavage site on fibronectin prevented the KLK12-dependent inhibition of fibronectin polymerization and the KLK12-mediated pro-migratory effect on endothelial cells. Taken as a whole, our results indicate that KLK12's proangiogenic effect is mediated through several molecular mechanisms.


Asunto(s)
Células Endoteliales/metabolismo , Fibronectinas/metabolismo , Calicreínas/metabolismo , Inductores de la Angiogénesis , Anticuerpos/metabolismo , Adhesión Celular/fisiología , Línea Celular , Movimiento Celular/fisiología , Matriz Extracelular/metabolismo , Humanos , Hidrólisis , Calicreínas/fisiología , Microvasos/metabolismo
6.
Artículo en Inglés | MEDLINE | ID: mdl-28078811

RESUMEN

The blood coagulation balance in the organism is achieved by the interaction of the blood platelets (PLTs) with the plasma coagulation system (PCS) and the vascular endothelial cells. In healthy organism, these systems prevent thrombosis and, in events of vascular damage, enable blood clotting to stop bleeding. The dysregulation of hemostasis may cause serious thrombotic and/or hemorrhagic pathologies. Numerous engineered nanomaterials are being investigated for biomedical purposes and are unavoidably exposed to the blood. Also, nanomaterials may access vascular system after occupational, environmental, or other types of exposure. Thus, it is essential to evaluate the effects of engineered nanomaterials on hemostasis. This review focuses on investigations of nanomaterial interactions with the blood components involved in blood coagulation: the PCS and PLTs. Particular emphases include the pathophysiology of effects of nanomaterials on the PCS, including the kallikrein-kinin system, and on PLTs. Methods for investigating these interactions are briefly described, and a review of the most important studies on the interactions of nanomaterials with plasma coagulation and platelets is provided. WIREs Nanomed Nanobiotechnol 2017, 9:e1448. doi: 10.1002/wnan.1448 For further resources related to this article, please visit the WIREs website.


Asunto(s)
Coagulación Sanguínea , Hemostasis , Nanoestructuras/uso terapéutico , Trombosis , Plaquetas/efectos de los fármacos , Humanos , Calicreínas/fisiología , Cininas/fisiología , Agregación Plaquetaria
7.
J Invest Dermatol ; 137(2): 449-456, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27769847

RESUMEN

Atopic dermatitis (AD) is a common inflammatory skin disorder. Chronic AD lesions present hyperkeratosis, indicating a disturbed desquamation process. KLK7 is a serine protease involved in the proteolysis of extracellular corneodesmosome components, including desmocollin 1 and corneodesmosin, which leads to desquamation. KLK7 is secreted by lamellar granules and upregulated in AD lesional skin. However, despite increased KLK7 protein levels, immunostaining and electron microscopy indicated numerous corneodesmosomes remaining in the uppermost layer of the stratum corneum from AD lesions. We aimed to clarify the discrepancy between KLK7 overexpression and retention of corneodesmosomes on AD corneocytes. Western blot analysis indicated abnormal corneodesmosin degradation patterns in stratum corneum from AD lesions. The KLK activity of tape-stripped corneocytes from AD lesions was not significantly elevated in in situ zymography, which was our new attempt to detect the protease activity more precisely than conventional assays. This ineffective KLK activation was associated with impaired KLK7 secretion from lamellar granules and increased expression of LEKTI in AD. Such imbalances in protease-protease inhibitor interactions could lead to abnormal proteolysis of corneodesmosomes and compact hyperkeratosis. Upregulated expression of LEKTI might be a compensatory mechanism to prevent further barrier dysfunction in AD.


Asunto(s)
Dermatitis Atópica/complicaciones , Epidermis/metabolismo , Calicreínas/fisiología , Queratosis/etiología , Proteínas Inhibidoras de Proteinasas Secretoras/fisiología , Desmosomas/metabolismo , Humanos , Inhibidor de Serinpeptidasas Tipo Kazal-5
8.
J Invest Dermatol ; 137(2): 377-384, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27765722

RESUMEN

Bacteria that reside on the skin can influence the behavior of the cutaneous immune system, but the mechanisms responsible for these effects are incompletely understood. Colonization of the skin by Staphylococcus aureus (S. aureus) is increased in atopic dermatitis and can result in increased severity of the disease. In this study, we show that S. aureus stimulates human keratinocytes to increase their endogenous protease activity, including specific increases in trypsin activity. This increased protease activity coincided with increased expression of mRNA for kallikreins (KLKs), with KLK6, 13, and 14 showing the greatest induction after exposure to S. aureus. Suppression of mRNA for these KLKs in keratinocytes by targeted small interfering RNA silencing before S. aureus exposure blocked the increase in protease activity. Keratinocytes exposed to S. aureus showed enhanced degradation of desmoglein-1 and filaggrin, whereas small interfering RNA for KLK6, KLK13, and KLK14 partially blocked this degradation. These data illustrate how S. aureus directly influences the skin barrier integrity by stimulating endogenous proteolytic activity and defines a previously unknown mechanism by which S. aureus may influence skin diseases.


Asunto(s)
Queratinocitos/enzimología , Serina Proteasas/metabolismo , Staphylococcus aureus/patogenicidad , Animales , Células Cultivadas , Desmogleína 1/metabolismo , Femenino , Proteínas Filagrina , Humanos , Proteínas de Filamentos Intermediarios/metabolismo , Calicreínas/fisiología , Ratones , Ratones Endogámicos C57BL
9.
Oncotarget ; 8(9): 14502-14515, 2017 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-27845893

RESUMEN

KLK6 is a serine protease highly expressed in the nervous system. In synucleinopathies, including Parkinson disease, the levels of KLK6 inversely correlate with α-synuclein in CSF. Recently, we suggested that recombinant KLK6 mediates the degradation of extracellular α-synuclein directly and via a proteolytic cascade that involves unidentified metalloproteinase(s). Here, we show that recombinant and naturally secreted KLK6 can readily cleave α-synuclein fibrils that have the potential for cell-to-cell propagation in "a prion-like mechanism". Importantly, KLK6-deficient primary cortical neurons have increased ability for α-synuclein fibril uptake. We also demonstrate that KLK6 activates proMMP2, which in turn can cleave α-synuclein. The repertoire of proteases activated by KLK6 in a neuronal environment was analyzed by degradomic profiling, which also identified ADAMTS19 and showed that KLK6 has a limited number of substrates indicating specific biological functions such as the regulation of α-synuclein turnover. We generated adenoviral vectors for KLK6 delivery and demonstrated that the levels of extracellular α-synuclein can be reduced by neuronally secreted KLK6. Our findings open the possibility to exploit KLK6 as a novel therapeutic target for Parkinson disease and other synucleinopathies.


Asunto(s)
Espacio Extracelular/metabolismo , Calicreínas/fisiología , Neuronas/metabolismo , Proteínas Quinasas/metabolismo , alfa-Sinucleína/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/citología , Proteolisis
10.
J Proteome Res ; 15(8): 2466-78, 2016 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-27378148

RESUMEN

Prostate cancer metastasis to bone is terminal; thus, novel therapies are required to prevent end-stage disease. Kallikrein-related peptidase 4 (KLK4) is a serine protease that is overproduced in localized prostate cancer and is abundant in prostate cancer bone metastases. In vitro, KLK4 induces tumor-promoting phenotypes; however, the underlying proteolytic mechanism is undefined. The protein topography and migration analysis platform (PROTOMAP) was used for high-depth identification of KLK4 substrates secreted by prostate cancer bone metastasis-derived PC-3 cells to delineate the mechanism of KLK4 action in advanced prostate cancer. Thirty-six putative novel substrates were determined from the PROTOMAP analysis. In addition, KLK4 cleaved the established substrate, urokinase-type plasminogen activator, thus validating the approach. KLK4 activated matrix metalloproteinase-1 (MMP1), a protease that promotes prostate tumor growth and metastasis. MMP1 was produced in the tumor compartment of prostate cancer bone metastases, highlighting its accessibility to KLK4 at this site. KLK4 further liberated an N-terminal product, with purported angiogenic activity, from thrombospondin-1 (TSP1) and cleaved TSP1 in an osteoblast-derived matrix. This is the most comprehensive analysis of the proteolytic action of KLK4 in an advanced prostate cancer model to date, highlighting KLK4 as a potential multifunctional regulator of prostate cancer progression.


Asunto(s)
Calicreínas/fisiología , Metaloproteinasa 1 de la Matriz/metabolismo , Neoplasias de la Próstata/patología , Trombospondina 1/metabolismo , Neoplasias Óseas/química , Neoplasias Óseas/metabolismo , Neoplasias Óseas/secundario , Progresión de la Enfermedad , Humanos , Masculino , Neoplasias de la Próstata/química , Proteolisis
11.
Urol Clin North Am ; 43(2): 209-16, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27132578

RESUMEN

Changes in understanding regarding the relationship of androgens and prostate cancer have led to changes in the use of testosterone therapy. The evidence supports a finite ability of androgens to stimulate prostate cancer growth, with a maximum achieved at low testosterone concentrations, called the saturation model. The saturation point corresponds with maximal androgenic stimulation at 250 ng/dL. Evidence is reviewed herein regarding the relationship of testosterone to prostate cancer and the relatively new practice of offering testosterone therapy to men with a history of prostate cancer. Although no prospective controlled trials have been performed, results have been reassuring.


Asunto(s)
Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/fisiopatología , Testosterona/fisiología , Testosterona/uso terapéutico , Humanos , Calicreínas/sangre , Calicreínas/fisiología , Masculino , Antígeno Prostático Específico/sangre , Antígeno Prostático Específico/fisiología , Neoplasias de la Próstata/sangre , Testosterona/sangre
12.
Exp Cell Res ; 333(1): 136-46, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25724897

RESUMEN

BACKGROUND: Ephrin-B2 is the sole physiologically-relevant ligand of the receptor tyrosine kinase EphB4, which is over-expressed in many epithelial cancers, including 66% of prostate cancers, and contributes to cancer cell survival, invasion and migration. Crucially, however, the cancer-promoting EphB4 signalling pathways are independent of interaction with its ligand ephrin-B2, as activation of ligand-dependent signalling causes tumour suppression. Ephrin-B2, however, is often found on the surface of endothelial cells of the tumour vasculature, where it can regulate angiogenesis to support tumour growth. Proteolytic cleavage of endothelial cell ephrin-B2 has previously been suggested as one mechanism whereby the interaction between tumour cell-expressed EphB4 and endothelial cell ephrin-B2 is regulated to support both cancer promotion and angiogenesis. METHODS: An in silico approach was used to search accessible surfaces of 3D protein models for cleavage sites for the key prostate cancer serine protease, KLK4, and this identified murine ephrin-B2 as a potential KLK4 substrate. Mouse ephrin-B2 was then confirmed as a KLK4 substrate by in vitro incubation of recombinant mouse ephrin-B2 with active recombinant human KLK4. Cleavage products were visualised by SDS-PAGE, silver staining and Western blot and confirmed by N-terminal sequencing. RESULTS: At low molar ratios, KLK4 cleaved murine ephrin-B2 but other prostate-specific KLK family members (KLK2 and KLK3/PSA) were less efficient, suggesting cleavage was KLK4-selective. The primary KLK4 cleavage site in murine ephrin-B2 was verified and shown to correspond to one of the in silico predicted sites between extracellular domain residues arginine 178 and asparagine 179. Surprisingly, the highly homologous human ephrin-B2 was poorly cleaved by KLK4 at these low molar ratios, likely due to the 3 amino acid differences at this primary cleavage site. CONCLUSION: These data suggest that in in vivo mouse xenograft models, endogenous mouse ephrin-B2, but not human tumour ephrin-B2, may be a downstream target of cancer cell secreted human KLK4. This is a critical consideration when interpreting data from murine explants of human EphB4+/KLK4+ cancer cells, such as prostate cancer cells, where differential effects may be seen in mouse models as opposed to human clinical situations.


Asunto(s)
Efrina-B2/química , Calicreínas/química , Calicreínas/metabolismo , Secuencia de Aminoácidos , Animales , Humanos , Calicreínas/fisiología , Masculino , Ratones , Datos de Secuencia Molecular , Trasplante de Neoplasias , Neoplasias de la Próstata , Proteolisis , Células Sf9
13.
Toxicol Appl Pharmacol ; 280(2): 216-23, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25110056

RESUMEN

Antineoplastic treatment with cisplatin is frequently complicated by nephrotoxicity. Although oxidative stress may be involved, the pathogenic mechanisms responsible for renal damage have not been completely clarified. In order to investigate the role of the renal kinin system in this condition, a group of rats was submitted to high potassium diet to stimulate the synthesis and excretion of tissue kallikrein 1 (rKLK1) previous to an intraperitoneal injection of 7 mg/kg cisplatin. A significant reduction in lipoperoxidation, evidenced by urinary excretion of malondialdehyde and renal immunostaining of hidroxy-nonenal, was accompanied by a decline in apoptosis. Coincident with these findings we observed a reduction in the expression of renal KIM-1 suggesting that renoprotection may be occurring. Stimulation or indemnity of the renal kinin system deserves to be evaluated as a complementary pharmacological measure to diminish cisplatin nephrotoxicity.


Asunto(s)
Lesión Renal Aguda/inducido químicamente , Antineoplásicos/toxicidad , Cisplatino/toxicidad , Calicreínas/fisiología , Animales , Moléculas de Adhesión Celular/análisis , Riñón/efectos de los fármacos , Masculino , Malondialdehído/orina , Ratas , Ratas Sprague-Dawley , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1
15.
Oncotarget ; 5(9): 2390-403, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24158494

RESUMEN

Kallikrein-related peptidase 5 (KLK5) displays aberrant expression in cancer. However, any functional association is missing. Here, we show that reconstitution of KLK5 expression in non-expressing MDA-MB-231 breast cancer cells suppresses malignancy in vitro and in vivo dose-dependently. Reactivation of KLK5 suppressed key EMT genes. Unexpectedly, we identified altered expression of genes encoding enzymes of the mevalonate pathway typical of those observed upon cholesterol starvation. Consistently, we found that SREBF1, the master regulator of the mevalonate pathway was induced. KLK5 re-expression leads to reduced cellular cholesterol and fatty acid synthesis and enhanced uptake of LDL-cholesterol. Suppression of the mevalonate pathway in KLK5 transfectants was further shown by reduced synthesis of isoprenoids. Indeed, we found diminished levels of active RhoA, a signaling oncoprotein that requires prenylation for activation. We propose that reduced RhoA activation plays a dominant role in suppression of malignancy by KLK5, since geranylgeranyl pyrophosphate restored active RhoA in KLK5-reverted cells resulting in increased malignancy. For the first time, we suggest that a protease may suppress breast cancer by modulating the mevalonate pathway.


Asunto(s)
Neoplasias de la Mama/prevención & control , Calicreínas/fisiología , Ácido Mevalónico/metabolismo , Transducción de Señal , Animales , Apoptosis , Western Blotting , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular , Colesterol/metabolismo , LDL-Colesterol/metabolismo , Ácidos Grasos/metabolismo , Femenino , Humanos , Técnicas para Inmunoenzimas , Ratones , Ratones SCID , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína de Unión al GTP rhoA/metabolismo
16.
Urology ; 82(6): 1197-203, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23992971

RESUMEN

The US diet has been fortified with folic acid to prevent neural tube defects since 1998. The Physician Data Queries from the National Cancer Institute describe folate as protective against prostate cancer, whereas its synthetic analog, folic acid, is considered to increase prostate cancer risk when taken at levels easily achievable by eating fortified food or taking over-the-counter supplements. We review the present literature to examine the effects of folate and folic acid on prostate cancer, help interpret previous epidemiologic data, and provide clarification regarding the apparently opposing roles of folate for patients with prostate cancer. A literature search was conducted in Medline to identify studies investigating the effect of nutrition and specifically folate and folic acid on prostate carcinogenesis and progression. In addition, the National Health and Nutrition Examination Survey database was analyzed for trends in serum folate levels before and after mandatory fortification. Folate likely plays a dual role in prostate carcinogenesis. There remains conflicting epidemiologic evidence regarding folate and prostate cancer risk; however, there is growing experimental evidence that higher circulating folate levels can contribute to prostate cancer progression. Further research is needed to clarify these complex relationships.


Asunto(s)
Neoplasias de la Próstata/fisiopatología , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Línea Celular Tumoral/metabolismo , Progresión de la Enfermedad , Ácido Fólico/sangre , Ácido Fólico/fisiología , Deficiencia de Ácido Fólico/epidemiología , Humanos , Inmunohistoquímica , Calicreínas/metabolismo , Calicreínas/fisiología , Masculino , Encuestas Nutricionales , Antígeno Prostático Específico/metabolismo , Antígeno Prostático Específico/fisiología , Neoplasias de la Próstata/epidemiología , Neoplasias de la Próstata/metabolismo , Complejo Vitamínico B/fisiología
17.
Proc Natl Acad Sci U S A ; 110(28): E2572-81, 2013 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-23798432

RESUMEN

The androgen receptor (AR) and the phosphoinositide 3-kinase (PI3K)/protein kinase B/mammalian target of rapamycin (mTOR) signaling are two of the major proliferative pathways in a number of tissues and are the main therapeutic targets in various disorders, including prostate cancer (PCa). Previous work has shown that there is reciprocal feedback regulation of PI3K and AR signaling in PCa, suggesting that cotargeting both pathways may enhance therapeutic efficacy. Here we show that proteins encoded by two androgen-regulated genes, kallikrein related peptidase 4 (KLK4) and promyelocytic leukemia zinc finger (PLZF), integrate optimal functioning of AR and mTOR signaling in PCa cells. KLK4 interacts with PLZF and decreases its stability. PLZF in turn interacts with AR and inhibits its function as a transcription factor. PLZF also activates expression of regulated in development and DNA damage responses 1, an inhibitor of mTORC1. Thus, a unique molecular switch is generated that regulates both AR and PI3K signaling. Consistently, KLK4 knockdown results in a significant decline in PCa cell proliferation in vitro and in vivo, decreases anchorage-independent growth, induces apoptosis, and dramatically sensitizes PCa cells to apoptosis-inducing agents. Furthermore, in vivo nanoliposomal KLK4 siRNA delivery in mice bearing PCa tumors results in profound remission. These results demonstrate that the activities of AR and mTOR pathways are maintained by KLK4, which may thus be a viable target for therapy.


Asunto(s)
Andrógenos/metabolismo , Calicreínas/fisiología , Neoplasias de la Próstata/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Muerte Celular , División Celular , Activación Enzimática , Fase G1 , Técnicas de Silenciamiento del Gen , Humanos , Calicreínas/genética , Masculino , Neoplasias de la Próstata/patología , Receptores Androgénicos/metabolismo , Proteínas Quinasas S6 Ribosómicas/metabolismo
18.
J Biol Chem ; 288(24): 17179-89, 2013 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-23629652

RESUMEN

Filaggrin protein is synthesized in the stratum granulosum of the skin and contributes to the formation of the human skin barrier. Profilaggrin is cleaved by proteolytic enzymes and converted to functional filaggrin, but its processing mechanism remains not fully elucidated. Kallikrein-related peptidase 5 (KLK5) is a major serine protease found in the skin, which is secreted from lamellar granules following its expression in the stratum granulosum and activated in the extracellular space of the stratum corneum. Here, we searched for profilaggrin-processing protease(s) by partial purification of epidermal extracts and found KLK5 as a possible candidate. We used high performance liquid chromatography coupled with electrospray tandem mass spectrometry to show that KLK5 cleaves profilaggrin. Furthermore, based on a proximity ligation assay, immunohistochemistry, and immunoelectron microscopy analysis, we reveal that KLK5 and profilaggrin co-localize in the stratum granulosum in human epidermis. KLK5 knockdown in normal cultured human epidermal keratinocytes resulted in higher levels of profilaggrin, indicating that KLK5 potentially functions in profilaggrin cleavage.


Asunto(s)
Proteínas de Filamentos Intermediarios/metabolismo , Calicreínas/fisiología , Proteolisis , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Células Cultivadas , Proteínas Filagrina , Técnicas de Silenciamiento del Gen , Humanos , Proteínas de Filamentos Intermediarios/química , Proteínas de Filamentos Intermediarios/genética , Calicreínas/química , Queratinocitos/enzimología , Ratones , Ratones Endogámicos C57BL , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , Transporte de Proteínas , ARN Interferente Pequeño/genética , Piel/citología , Piel/enzimología
19.
Biomarkers ; 18(4): 279-96, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23672534

RESUMEN

The kinin-kallikrein system (KKS) is an endogenous multiprotein cascade, the activation of which leads to triggering of the intrinsic coagulation pathway and enzymatic hydrolysis of kininogens with the consequent release of bradykinin-related peptides. This system plays a crucial role in inflammation, vasodilation, smooth muscle contraction, cardioprotection, vascular permeability, blood pressure control, coagulation and pain. In this review, we will outline the physiology and pathophysiology of the KKS and also highlight the association of this system with carcinogenesis and cancer progression.


Asunto(s)
Biomarcadores de Tumor/fisiología , Calicreínas/fisiología , Cininas/metabolismo , Humanos
20.
J Neurochem ; 127(2): 283-98, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23647384

RESUMEN

CNS trauma generates a proteolytic imbalance contributing to secondary injury, including axonopathy and neuron degeneration. Kallikrein 6 (Klk6) is a serine protease implicated in neurodegeneration, and here we investigate the role of protease-activated receptors 1 (PAR1) and PAR2 in mediating these effects. First, we demonstrate Klk6 and the prototypical activator of PAR1, thrombin, as well as PAR1 and PAR2, are each elevated in murine experimental traumatic spinal cord injury (SCI) at acute or subacute time points. Recombinant Klk6 triggered extracellular signal-regulated kinase (ERK1/2) signaling in cerebellar granule neurons and in the NSC34 spinal cord motoneuron cell line, in a phosphoinositide 3-kinae and MEK-dependent fashion. Importantly, lipopeptide inhibitors of PAR1 or PAR2, and PAR1 genetic deletion, each reduced Klk6-ERK1/2 activation. In addition, Klk6 and thrombin promoted degeneration of cerebellar neurons and exacerbated glutamate neurotoxicity. Moreover, genetic deletion of PAR1 blocked thrombin-mediated cerebellar neurotoxicity and reduced the neurotoxic effects of Klk6. Klk6 also increased glutamate-mediated Bim signaling, poly-ADP-ribose polymerase cleavage and lactate dehydrogenase release in NSC34 motoneurons and these effects were blocked by PAR1 and PAR2 lipopeptide inhibitors. Taken together, these data point to a novel Klk6-signaling axis in CNS neurons that is mediated by PAR1 and PAR2 and is positioned to contribute to neurodegeneration.


Asunto(s)
Ácido Glutámico/toxicidad , Calicreínas/fisiología , Neuronas/patología , Síndromes de Neurotoxicidad/fisiopatología , Receptor PAR-1/fisiología , Receptor PAR-2/fisiología , Animales , Células Cultivadas , Cerebelo/citología , Femenino , Calicreínas/genética , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas Motoras/fisiología , ARN/biosíntesis , ARN/genética , Receptor PAR-1/genética , Receptor PAR-2/genética , Transducción de Señal/fisiología , Médula Espinal/citología , Médula Espinal/fisiología , Traumatismos de la Médula Espinal/patología , Trombina/genética , Trombina/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...