Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 252
Filtrar
1.
Analyst ; 148(23): 5926-5934, 2023 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-37850419

RESUMEN

Angiotensin and kinin metabolic pathways are reported to be altered by many diseases, including COVID-19. Monitoring levels of these peptide metabolites is important for understanding mechanisms of disease processes. In this paper, we report dimethyl labeling of amines in peptides by addition of formaldehyde to samples and deutero-formaldehyde to internal standards to generate nearly identical isotopic standards with 4 m/z units larger per amine group than the corresponding analyte. We apply this approach to rapid, multiplexed, absolute LC-MS/MS quantitation of renin angiotensin system (RAS) and kallikrein-kinin system (KKS) peptides in human blood serum. Limits of detection (LODs) were obtained in the low pg mL-1 range with 3 orders of magnitude dynamic ranges, appropriate for determinations of normal and elevated levels of the target peptides in blood serum and plasma. Accuracy is within ±15% at concentrations above the limit of quantitation, as validated by spike-recovery in serum samples. Applicability was demonstrated by measuring RAS and KKS peptides in serum from COVID-19 patients, but is extendable to any class of peptides or other small molecules bearing reactive -NH2 groups.


Asunto(s)
COVID-19 , Sistema Renina-Angiotensina , Humanos , Sistema Calicreína-Quinina , Cromatografía Liquida , Suero , COVID-19/diagnóstico , Espectrometría de Masas en Tándem , Péptidos , Formaldehído , Isótopos
2.
J Hazard Mater ; 458: 132044, 2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37451104

RESUMEN

Atmospheric particulate matter (PM) perturbs hematological homeostasis by targeting the plasma kallikrein-kinin system (KKS), causing a cascade of zymogen activation events. However, the causative components involved in PM-induced hematological effects are largely unknown. Herein, the standard reference materials (SRMs) of atmospheric PM, including emissions from the diesel (2975), urban (1648a), and bituminous coal (2693), were screened for their effects on plasma KKS activation, and the effective constituent contributing to PM-induced KKS activation was further explored by fraction isolation and chemical analysis. The effects of three SRMs on KKS activation followed the order of 2975 > 1648a > 2693, wherein the fractions of 2975 isolated by acetone and water, together with the insoluble particulate residues, exerted significant perturbations in the hematological homeostasis. The soot contents in the SRMs and corresponding isolated fractions matched well with their hematological effects, and the KKS activation could be dependent on the soot surface oxidation degree. This study, for the first time, uncovered the soot content in atmospheric PM with different origins contributed to the distinct effects on plasma KKS activation. The finding would be of utmost importance for the health risk assessment on inhaled airborne fine PM, given its inevitable contact with human circulatory system.


Asunto(s)
Contaminantes Atmosféricos , Sistema Calicreína-Quinina , Material Particulado , Humanos , Sistema Calicreína-Quinina/fisiología , Hollín , Contaminantes Atmosféricos/análisis
3.
Am J Physiol Cell Physiol ; 324(3): C741-C756, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36745527

RESUMEN

Vasoactive peptides often serve a multitude of functions aside from their direct effects on vasodynamics. This article will review the existing literature on two vasoactive peptides and their involvement in skin homeostasis: adiponectin and-as the main representative of the kallikrein-kinin system-bradykinin. Adiponectin is the most abundantly expressed adipokine in the human organism, where it is mainly localized in fat depots including subcutaneous adipose tissue, from where adiponectin can exert paracrine effects. The involvement of adiponectin in skin homeostasis is supported by a number of studies reporting the effects of adiponectin in isolated human keratinocytes, sebocytes, fibroblasts, melanocytes, and immune cells. Regarding skin pathology, the potential involvement of adiponectin in psoriasis, atopic dermatitis, scleroderma, keloid, and melanogenesis is discussed in this article. The kallikrein-kinin system is composed of a variety of enzymes and peptides, most of which have been identified to be expressed in the skin. This also includes the expression of bradykinin receptors on most skin cells. Bradykinin is one of the very few hormones that is targeted by treatment in routine clinical use in dermatology-in this case for the treatment of hereditary angioedema. The potential involvement of bradykinin in wound healing, psoriasis, and melanoma is further discussed in this article. This review concludes with a call for additional preclinical and clinical studies to further explore the therapeutic potential of adiponectin supplementation (for psoriasis, atopic dermatitis, wound healing, scleroderma, and keloid) or pharmacological interference with the kallikrein-kinin system (for wound healing, psoriasis, and melanoma).


Asunto(s)
Adiponectina , Bradiquinina , Homeostasis , Sistema Calicreína-Quinina , Enfermedades de la Piel , Fenómenos Fisiológicos de la Piel , Adiponectina/fisiología , Sistema Calicreína-Quinina/fisiología , Bradiquinina/fisiología , Humanos , Enfermedades de la Piel/metabolismo
4.
Int J Mol Sci ; 23(18)2022 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-36142208

RESUMEN

Rheumatoid arthritis is an autoimmune disease that affects joints, leading to swelling, inflammation, and dysfunction in the joints. Recently, research efforts have been focused on finding novel curative approaches for rheumatoid arthritis, as current therapies are associated with adverse effects. Here, we examined the effectiveness of dabigatran, the antithrombotic agent, in treating complete Freund's adjuvant (CFA)-induced arthritis in rats. Subcutaneous injection of a single 0.3 mL dosage of CFA into the rat's hind leg planter surface resulted in articular surface deformities, reduced cartilage thickness, loss of intercellular matrix, and inflammatory cell infiltration. There were also increased levels of the Anti-cyclic citrullinated peptide antibody (ACPA), oxidative stress, and tissue Receptor activator of nuclear factor-kappa B ligand (RANKL). Proteins of the kallikrein-kinin system (KKS) were also elevated. The inhibitory effects of dabigatran on thrombin led to a subsequent inhibition of KKS and reduced Toll-like receptor 4 (TLR4) expression. These effects also decreased RANKL levels and showed anti-inflammatory and antioxidant effects. Therefore, dabigatran could be a novel therapeutic strategy for arthritis.


Asunto(s)
Artritis Experimental , Artritis Reumatoide , Animales , Antiinflamatorios/efectos adversos , Antioxidantes/metabolismo , Artritis Experimental/metabolismo , Artritis Reumatoide/inducido químicamente , Artritis Reumatoide/tratamiento farmacológico , Dabigatrán/farmacología , Dabigatrán/uso terapéutico , Fibrinolíticos/uso terapéutico , Adyuvante de Freund/efectos adversos , Sistema Calicreína-Quinina , Ligando RANK/metabolismo , Ratas , Trombina/metabolismo , Receptor Toll-Like 4/metabolismo
5.
Sci Rep ; 12(1): 14167, 2022 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-35986069

RESUMEN

Heart transplantation remains the definitive treatment for end stage heart failure. Because availability is limited, risk stratification of candidates is crucial for optimizing both organ allocations and transplant outcomes. Here we utilize proteomics prior to transplant to identify new biomarkers that predict post-transplant survival in a multi-institutional cohort. Microvesicles were isolated from serum samples and underwent proteomic analysis using mass spectrometry. Monte Carlo cross-validation (MCCV) was used to predict survival after transplant incorporating select recipient pre-transplant clinical characteristics and serum microvesicle proteomic data. We identified six protein markers with prediction performance above AUROC of 0.6, including Prothrombin (F2), anti-plasmin (SERPINF2), Factor IX, carboxypeptidase 2 (CPB2), HGF activator (HGFAC) and low molecular weight kininogen (LK). No clinical characteristics demonstrated an AUROC > 0.6. Putative biological functions and pathways were assessed using gene set enrichment analysis (GSEA). Differential expression analysis identified enriched pathways prior to transplant that were associated with post-transplant survival including activation of platelets and the coagulation pathway prior to transplant. Specifically, upregulation of coagulation cascade components of the kallikrein-kinin system (KKS) and downregulation of kininogen prior to transplant were associated with survival after transplant. Further prospective studies are warranted to determine if alterations in the KKS contributes to overall post-transplant survival.


Asunto(s)
Trasplante de Corazón , Sistema Calicreína-Quinina , Coagulación Sanguínea , Trasplante de Corazón/efectos adversos , Humanos , Sistema Calicreína-Quinina/fisiología , Quininógenos/metabolismo , Proteómica
6.
Front Immunol ; 12: 767347, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34804054

RESUMEN

Infection with SARS-CoV-2 triggers the simultaneous activation of innate inflammatory pathways including the complement system and the kallikrein-kinin system (KKS) generating in the process potent vasoactive peptides that contribute to severe acute respiratory syndrome (SARS) and multi-organ failure. The genome of SARS-CoV-2 encodes four major structural proteins - the spike (S) protein, nucleocapsid (N) protein, membrane (M) protein, and the envelope (E) protein. However, the role of these proteins in either binding to or activation of the complement system and/or the KKS is still incompletely understood. In these studies, we used: solid phase ELISA, hemolytic assay and surface plasmon resonance (SPR) techniques to examine if recombinant proteins corresponding to S1, N, M and E: (a) bind to C1q, gC1qR, FXII and high molecular weight kininogen (HK), and (b) activate complement and/or the KKS. Our data show that the viral proteins: (a) bind C1q and activate the classical pathway of complement, (b) bind FXII and HK, and activate the KKS in normal human plasma to generate bradykinin and (c) bind to gC1qR, the receptor for the globular heads of C1q (gC1q) which in turn could serve as a platform for the activation of both the complement system and KKS. Collectively, our data indicate that the SARS-CoV-2 viral particle can independently activate major innate inflammatory pathways for maximal damage and efficiency. Therefore, if efficient therapeutic modalities for the treatment of COVID-19 are to be designed, a strategy that includes blockade of the four major structural proteins may provide the best option.


Asunto(s)
Antígenos Virales/inmunología , COVID-19/inmunología , Proteínas del Sistema Complemento/inmunología , Sistema Calicreína-Quinina , SARS-CoV-2/inmunología , Proteínas Estructurales Virales/inmunología , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Hemólisis , Humanos , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/inmunología , Proteínas Recombinantes/inmunología , Proteínas Estructurales Virales/genética
7.
Molecules ; 26(14)2021 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-34299400

RESUMEN

The goal of this study was to assess the pharmacological effects of black tea (Camellia sinensis var. assamica) water extract on human kinin-forming enzymes in vitro. Tea is a highly consumed beverage in the world. Factor XII (FXII, Hageman factor)-independent- and -dependent activation of prekallikrein to kallikrein leads to the liberation of bradykinin (BK) from high-molecular-weight kininogen (HK). The excessive BK production causes vascular endothelial and nonvascular smooth muscle cell permeability, leading to angioedema. The prevalence of angiotensin-converting enzyme inhibitor (ACEI)-induced angioedema appears to be through BK. Both histamine and BK are potent inflammatory mediators. However, the treatments for histamine-mediated angioedema are unsuitable for BK-mediated angioedema. We hypothesized that long-term consumption of tea would reduce bradykinin-dependent processes within the systemic and pulmonary vasculature, independent of the anti-inflammatory actions of polyphenols. A purified fraction of the black tea water extract inhibited both kallikrein and activated FXII. The black tea water extracts inhibited factor XII-induced cell migration and inhibited the production of kallikrein on the endothelial cell line. We compared the inhibitory effects of the black tea water extract and twenty-three well-known anti-inflammatory medicinal herbs, in inhibiting both kallikrein and FXII. Surprisingly, arjunglucoside II specifically inhibited the activated factor XII (FXIIa), but not the kallikrein and the activated factor XI. Taken together, the black tea water extract exerts its anti-inflammatory effects, in part, by inhibiting kallikrein and activated FXII, which are part of the plasma kallikrein-kinin system (KKS), and by decreasing BK production. The inhibition of kallikrein and activated FXII represents a unique polyphenol-independent anti-inflammatory mechanism of action for the black tea.


Asunto(s)
Bradiquinina/metabolismo , Camellia/química , Endotelio Vascular/efectos de los fármacos , Factor XII/antagonistas & inhibidores , Sistema Calicreína-Quinina/efectos de los fármacos , Extractos Vegetales/farmacología , Arteria Pulmonar/efectos de los fármacos , Proliferación Celular , Células Cultivadas , Endotelio Vascular/metabolismo , Humanos , Arteria Pulmonar/metabolismo
9.
Semin Immunopathol ; 43(4): 507-517, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34125270

RESUMEN

Coagulation is controlled by a delicate balance of prothrombotic and antithrombotic mechanisms, to prevent both excessive blood loss from injured vessels and pathologic thrombosis. The liver plays a pivotal role in hemostasis through the synthesis of plasma coagulation factors and their inhibitors that, in addition to thrombosis and hemostasis, orchestrates an array of inflammatory responses. As a result, impaired liver function has been linked with both hypercoagulability and bleeding disorders due to a pathologic balance of pro- and anticoagulant plasma factors. At sites of vascular injury, thrombus propagation that finally may occlude the blood vessel depends on negatively charged biopolymers, such as polyphosphates and extracellular DNA, that provide a physiological surface for contact activation of coagulation factor XII (FXII). FXII initiates the contact system that drives both the intrinsic pathway of coagulation, and formation of the inflammatory mediator bradykinin by the kallikrein-kinin system. Moreover, FXII facilitates receptor-mediated signalling, thereby promoting mitogenic activities, angiogenesis, and neutrophil stimulation with implications for liver diseases. Here, we summarize current knowledge on the FXII-driven contact system in liver diseases and review therapeutic approaches to target its activities during impaired liver function.


Asunto(s)
Factor XII , Trombosis , Coagulación Sanguínea , Factor XII/metabolismo , Humanos , Sistema Calicreína-Quinina , Hígado/metabolismo , Trombosis/etiología
10.
J Investig Allergol Clin Immunol ; 31(1): 1-16, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33602658

RESUMEN

Hereditary angioedema due to C1-esterase inhibitor deficiency (C1-INH-HAE) is a rare autosomal dominant disease. In the last decade, new drugs and new indications for old drugs have played a role in the management of C1-INH-HAE. This review examines current therapy for C1-INH-HAE and provides a brief summary of drugs that are under development. Increased knowledge of the pathophysiology of C1-INH-HAE has been crucial for advances in the field, with inhibition of the kallikrein-kinin system (plasma kallikrein, activated factor XII) as a key area in the discovery of new drugs, some of which are already marketed for treatment of C1-INH-HAE. Pharmacological treatment is based on 3 pillars: treatment of acute angioedema attacks (on-demand treatment), short-term (preprocedure) prophylaxis, and long-term prophylaxis. The 4 drugs that are currently available for the treatment of acute angioedema attacks (purified plasma-derived human C1 esterase inhibitor concentrate, icatibant acetate, ecallantide, recombinant human C1 esterase inhibitor) are all authorized for self-administration, except ecallantide. Purified plasma-derived human C1 esterase inhibitor concentrate is the treatment of choice for short-term prophylaxis. Tranexamic acid, danazol, intravenous and subcutaneous nanofiltered purified plasma-derived human C1 esterase inhibitor concentrate, and lanadelumab can be used for long-term prophylaxis. New drugs are being investigated, mainly as long-term prophylaxis, and are aimed at blocking the kallikrein-kinin system by means of antiprekallikrein, antikallikrein, and anti-activated FXII action.


Asunto(s)
Angioedemas Hereditarios/tratamiento farmacológico , Antiinflamatorios no Esteroideos/uso terapéutico , Bradiquinina/análogos & derivados , Proteína Inhibidora del Complemento C1/uso terapéutico , Péptidos/uso terapéutico , Angioedemas Hereditarios/genética , Bradiquinina/uso terapéutico , Proteína Inhibidora del Complemento C1/genética , Humanos , Sistema Calicreína-Quinina , Proteínas Recombinantes/genética
11.
J Renin Angiotensin Aldosterone Syst ; 21(4): 1470320320979097, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33283602

RESUMEN

Because of the current COVID-19-pandemic, the world is currently being held hostage in various lockdowns. ACE2 facilitates SARS-CoV-2 cell-entry, and is at the very center of several pathophysiological pathways regarding the RAAS, CS, KKS, T2DM, and IL-6. Their interactions with severe COVID-19 complications (e.g. ARDS and thrombosis), and potential therapeutic targets for pharmacological intervention, will be reviewed.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/metabolismo , Proteínas del Sistema Complemento/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/virología , Interleucina-6/metabolismo , Sistema Calicreína-Quinina , COVID-19/complicaciones , Diabetes Mellitus Tipo 2/complicaciones , Humanos , SARS-CoV-2/fisiología , Tratamiento Farmacológico de COVID-19
12.
Int J Mol Sci ; 21(21)2020 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-33138181

RESUMEN

The 1918 influenza killed approximately 50 million people in a few short years, and now, the world is facing another pandemic. In December 2019, a novel coronavirus named severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused an international outbreak of a respiratory illness termed coronavirus disease 2019 (COVID-19) and rapidly spread to cause the worst pandemic since 1918. Recent clinical reports highlight an atypical presentation of acute respiratory distress syndrome (ARDS) in COVID-19 patients characterized by severe hypoxemia, an imbalance of the renin-angiotensin system, an increase in thrombogenic processes, and a cytokine release storm. These processes not only exacerbate lung injury but can also promote pulmonary vascular remodeling and vasoconstriction, which are hallmarks of pulmonary hypertension (PH). PH is a complication of ARDS that has received little attention; thus, we hypothesize that PH in COVID-19-induced ARDS represents an important target for disease amelioration. The mechanisms that can promote PH following SARS-CoV-2 infection are described. In this review article, we outline emerging mechanisms of pulmonary vascular dysfunction and outline potential treatment options that have been clinically tested.


Asunto(s)
Lesión Pulmonar Aguda/patología , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/patología , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/patología , Síndrome Respiratorio Agudo Grave/patología , Vasoconstricción/fisiología , Betacoronavirus , COVID-19 , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Síndrome de Liberación de Citoquinas/patología , Sistema Calicreína-Quinina/fisiología , Pandemias , Sistema Renina-Angiotensina/fisiología , SARS-CoV-2 , Síndrome Respiratorio Agudo Grave/tratamiento farmacológico , Vasoconstricción/efectos de los fármacos
13.
Pharmacol Res ; 160: 105096, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32712319

RESUMEN

The risk of thrombosis, a globally growing challenge and a major cause of death, is influenced by various factors in the intravascular coagulation, vessel wall, and cellular systems. Among the contributors to thrombosis, the contact activation system and the kallikrein/kinin system, two overlapping plasma proteolytic systems that are often considered as synonymous, regulate thrombosis from different aspects. On one hand, components of the contact activation system such as factor XII initiates activation of the coagulation proteins promoting thrombus formation on artificial surfaces through factor XI- and possibly prekallikrein-mediated intrinsic coagulation. On the other hand, physiological activation of plasma prekallikrein in the kallikrein/kinin system on endothelial cells liberates bradykinin from associated high-molecular-weight kininogen to stimulate the constitutive bradykinin B2 receptor to generate nitric oxide and prostacyclin to induce vasodilation and counterbalance angiotensin II signaling from the renin-angiotensin system which stimulates vasoconstriction. In addition to vascular tone regulation, this interaction between the kallikrein/kinin and renin-angiotensin systems has a thrombo-regulatory role independent of the contact pathway. At the level of the G-protein coupled receptors of these systems, defective bradykinin signaling due to attenuated bradykinin formation and/or decreased B2 receptor expression, as seen in murine prekallikrein and B2 receptor null mice, respectively, leads to compensatory overexpressed Mas, the receptor for angiotensin-(1-7) of the renin-angiotensin system. Mas stimulation and/or its increased expression contributes to maintaining a healthy vascular homeostasis by generating graded elevation of plasma prostacyclin which reduces thrombosis through two independent pathways: (1) increasing the vasoprotective transcription factor Sirtuin 1 to suppress tissue factor expression, and (2) inhibiting platelet activation. This review will summarize the recent advances in this field that support these understandings. Appreciating these subtle mechanisms help to develop novel anti-thrombotic strategies by targeting the vascular receptors in the renin-angiotensin and the kallikrein/kinin systems to maintain healthy vascular homeostasis.


Asunto(s)
Coagulación Sanguínea , Sistema Calicreína-Quinina/efectos de los fármacos , Proteínas Proto-Oncogénicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sistema Renina-Angiotensina , Trombosis/sangre , Animales , Epoprostenol/metabolismo , Humanos , Precalicreína/metabolismo , Proto-Oncogenes Mas , Receptor de Bradiquinina B2/metabolismo , Transducción de Señal , Sirtuina 1/metabolismo
14.
Int J Mol Sci ; 21(9)2020 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-32397357

RESUMEN

Recent studies have shown that the innate and adaptive immune system, together with low-grade inflammation, may play an important role in essential hypertension. In this work, to verify the importance of selected factors for the development of essential hypertension, we created a Petri net-based model and analyzed it. The analysis was based mainly on t-invariants, knockouts of selected fragments of the net and its simulations. The blockade of the renin-angiotensin (RAA) system revealed that the most significant effect on the emergence of essential hypertension has RAA activation. This blockade affects: (1) the formation of angiotensin II, (2) inflammatory process (by influencing C-reactive protein (CRP)), (3) the initiation of blood coagulation, (4) bradykinin generation via the kallikrein-kinin system, (5) activation of lymphocytes in hypertension, (6) the participation of TNF alpha in the activation of the acute phase response, and (7) activation of NADPH oxidase-a key enzyme of oxidative stress. On the other hand, we found that the blockade of the activation of the RAA system may not eliminate hypertension that can occur due to disturbances associated with the osmotically independent binding of Na in the interstitium. Moreover, we revealed that inflammation alone is not enough to trigger primary hypertension, but it can coexist with it. We believe that our research may contribute to a better understanding of the pathology of hypertension. It can help identify potential subprocesses, which blocking will allow better control of essential hypertension.


Asunto(s)
Hipertensión Esencial/fisiopatología , Inflamación/fisiopatología , Modelos Biológicos , Angiotensina II/fisiología , Autoantígenos/inmunología , Coagulación Sanguínea , Bradiquinina/biosíntesis , Proteína C-Reactiva/fisiología , Endotelio Vascular/inmunología , Hipertensión Esencial/etiología , Hipertensión Esencial/inmunología , Humanos , Inflamación/inmunología , Sistema Calicreína-Quinina/fisiología , Activación de Linfocitos , NADPH Oxidasas/fisiología , Natriuresis/fisiología , Óxido Nítrico/fisiología , Óxido Nítrico Sintasa de Tipo III/fisiología , Sistema Renina-Angiotensina/efectos de los fármacos , Sistema Renina-Angiotensina/fisiología , Piel/fisiopatología , Sodio/metabolismo , Cloruro de Sodio Dietético/farmacocinética , Factor de Necrosis Tumoral alfa/fisiología
15.
Acta Pharmacol Sin ; 41(12): 1597-1608, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32300244

RESUMEN

Tissue kallikrein has protective function against various types of injury. In this study, we investigated whether exogenous pancreatic kininogenase (PK) conferred renoprotection in a rat model of unilateral ureteral obstruction (UUO) and H2O2-treated HK-2 cells in vitro. SD rats were subjected to UUO surgery, then PK (7.2 U/g per day, ip) was administered for 7 or 14 days. After the treatment, rats were euthanized; the obstructed kidneys were harvested for further examination. We found that PK administration significantly attenuated interstitial inflammation and fibrosis, and downregulated the expression of proinflammatory (MCP-1, TLR-2, and OPN) and profibrotic (TGF-ß1 and CTGF) cytokines in obstructed kidney. UUO-induced oxidative stress, closely associated with excessive apoptotic cell death and autophagy via PI3K/AKT/FoxO1a signaling, which were abolished by PK administration. We further showed that PK administration increased the expression of bradykinin receptors 1 and 2 (B1R and B2R) mRNA and the production of NO and cAMP in kidney tissues. Coadministration with either B1R antagonist (des-Arg9-[Leu8]-bradykinin) or B2R antagonist (icatibant) abrogated the renoprotective effects of PK, and reduced the levels of NO and cAMP in obstructed kidney. In H2O2-treated HK-2 cells, addition of PK (6 pg/mL) significantly decreased ROS production, regulated the expression of oxidant and antioxidant enzymes, suppressed the expression of TGF-ß1 and MCP-1, and inhibited cell apoptosis. Our data demonstrate that PK treatment protects against the progression of renal fibrosis in obstructed kidneys.


Asunto(s)
Fibrosis/prevención & control , Calicreínas/uso terapéutico , Riñón/metabolismo , Páncreas/enzimología , Sustancias Protectoras/uso terapéutico , Obstrucción Ureteral/complicaciones , Animales , Muerte Celular/efectos de los fármacos , Línea Celular , Fibrosis/etiología , Fibrosis/patología , Humanos , Inflamación/tratamiento farmacológico , Inflamación/etiología , Inflamación/patología , Sistema Calicreína-Quinina/efectos de los fármacos , Riñón/patología , Masculino , Estrés Oxidativo/efectos de los fármacos , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Obstrucción Ureteral/patología
16.
Reprod Toxicol ; 93: 1-9, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31874189

RESUMEN

Kallikrein-kinin system (KKS) is involved in vascular reactivity and inflammatory response to cytotoxic drugs. Since cisplatin is a widely used chemotherapy and its cytotoxic mechanism can trigger inflammation and oxidative damage, in this work we evaluated the role of KKS in an animal model of cisplatin-induced ovarian toxicity. Biomarkers of ovarian stem cells, activity of KKS, inflammation and oxidative damage were measured in ovarian tissue of C57BL/6 female mice treated with vehicle or cisplatin (2.5 mg/kg). Cisplatin group presented greater number of atretic follicles, and lower numbers of antral and total viable follicles. Ki67, DDX4 and OCT-4 markers were similar between groups. Cisplatin triggered plasma and ovarian tissue kallikrein generation; and increased expression of bradykinin receptors B1 and B2. Neutrophil and macrophage infiltration markers increased. Superoxide anion generation also increased, while reduced glutathione levels decreased. These results suggest that KKS is activated and contributes to ovarian injury during cisplatin treatment.


Asunto(s)
Antineoplásicos/efectos adversos , Cisplatino/efectos adversos , Ovario/efectos de los fármacos , Animales , Femenino , Sistema Calicreína-Quinina , Calicreínas/metabolismo , Ratones Endogámicos C57BL , Óxido Nítrico/metabolismo , Ovario/metabolismo , Ovario/patología , Estrés Oxidativo/efectos de los fármacos , Receptor de Bradiquinina B1/metabolismo , Receptor de Bradiquinina B2/metabolismo
17.
Sci Rep ; 9(1): 19437, 2019 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-31857655

RESUMEN

Pharmacological research in mice and human genetic analyses suggest that the kallikrein-kinin system (KKS) may regulate anxiety. We examined the role of the KKS in anxiety and stress in both species. In human genetic association analysis, variants in genes for the bradykinin precursor (KNG1) and the bradykinin receptors (BDKRB1 and BDKRB2) were associated with anxiety disorders (p < 0.05). In mice, however, neither acute nor chronic stress affected B1 receptor gene or protein expression, and B1 receptor antagonists had no effect on anxiety tests measuring approach-avoidance conflict. We thus focused on the B2 receptor and found that mice injected with the B2 antagonist WIN 64338 had lowered levels of a physiological anxiety measure, the stress-induced hyperthermia (SIH), vs controls. In the brown adipose tissue, a major thermoregulator, WIN 64338 increased expression of the mitochondrial regulator Pgc1a and the bradykinin precursor gene Kng2 was upregulated after cold stress. Our data suggests that the bradykinin system modulates a variety of stress responses through B2 receptor-mediated effects, but systemic antagonists of the B2 receptor were not anxiolytic in mice. Genetic variants in the bradykinin receptor genes may predispose to anxiety disorders in humans by affecting their function.


Asunto(s)
Trastornos de Ansiedad/metabolismo , Bradiquinina/metabolismo , Sistema Calicreína-Quinina/fisiología , Estrés Psicológico/metabolismo , Adulto , Animales , Trastornos de Ansiedad/tratamiento farmacológico , Trastornos de Ansiedad/genética , Trastornos de Ansiedad/patología , Antagonistas del Receptor de Bradiquinina B1/administración & dosificación , Antagonistas del Receptor de Bradiquinina B2/administración & dosificación , Encéfalo/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Sistema Calicreína-Quinina/efectos de los fármacos , Quininógenos/genética , Quininógenos/metabolismo , Masculino , Ratones , Naftalenos/administración & dosificación , Compuestos Organofosforados/administración & dosificación , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Polimorfismo de Nucleótido Simple , Receptor de Bradiquinina B1/genética , Receptor de Bradiquinina B1/metabolismo , Receptor de Bradiquinina B2/genética , Receptor de Bradiquinina B2/metabolismo , Especificidad de la Especie , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/patología , Regulación hacia Arriba
18.
J Neurochem ; 150(3): 296-311, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31206169

RESUMEN

Temporal lobe epilepsy (TLE) is a chronic disease, characterized by severe and refractory seizures, triggered in the hippocampus and/or amygdala, disrupting the blood-brain barrier. This disruption can sustain, or aggravate, the epileptic condition. The aim of this study was to evaluate the activation of the kallikrein-kinin system in patients with TLE, as it relates to the maintenance of blood-brain barrier. Human hippocampal sclerotic tissues removed after surgery for seizure control, plasma, and serum were used in the following assays: immunostaining for white blood cells in the TLE hippocampus, C-reactive protein in serum, quantification of plasma kallikrein (PKal) and cathepsin B (CatB) activity in serum and plasma, quantification of C1-inhibitor, analysis of high-molecular-weight kininogen (H-kininogen) fragments, and activation of plasma prekallikrein for comparison with healthy controls. Infiltration of white blood cells in the sclerotic hippocampus and a significant increase in the neutrophil/lymphocyte ratio in the blood of TLE patients were observed. High levels of C-reactive protein (TLE = 1.4 ± 0.3 µg/mL), PKal (TLE = 5.4 ± 0.4 U/mL), and CatB (TLE = 4.9 ± 0.4 U/mL) were also evident in the serum of TLE patients comparing to controls. A strong linear correlation was observed between active CatB and PKal in the serum of TLE patients (r = 0.88). High levels of cleaved H-kininogen and free PKal, and low levels of C1-inhibitor (TLE = 188 ± 12 µg/mL) were observed in the serum of TLE patients. Our data demonstrated that the plasma kallikrein-kinin system is activated in patients with TLE. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.


Asunto(s)
Catepsina B/sangre , Epilepsia del Lóbulo Temporal/metabolismo , Inflamación/metabolismo , Sistema Calicreína-Quinina/fisiología , Calicreínas/sangre , Adulto , Femenino , Hipocampo/metabolismo , Humanos , Masculino , Persona de Mediana Edad
19.
JCI Insight ; 3(9)2018 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-29720566

RESUMEN

Kidney injury is a frequent outcome in patients with disseminated Candida albicans fungal infections. IL-17 receptor (IL-17R) signaling is critical for renal protection against disseminated candidiasis, but the identity and function of IL-17-responsive cells in mediating renal defense remains an active area of debate. Using BM chimeras, we found that IL-17R signaling is required only in nonhematopoietic cells for immunity to systemic C. albicans infection. Since renal tubular epithelial cells (RTEC) are highly responsive to IL-17 in vitro, we hypothesized that RTEC might be the dominant target of IL-17 activity in the infected kidney. We generated mice with a conditional deletion of IL-17 receptor A (Il17ra) in RTEC (Il17raΔRTEC). Strikingly, Il17raΔRTEC mice showed enhanced kidney damage and early mortality following systemic infection, very similar to Il17ra-/- animals. Increased susceptibility to candidiasis in Il17raΔRTEC mice was associated with diminished activation of the renal protective Kallikrein-kinin system (KKS), resulting in reduced apoptosis of kidney-resident cells during hyphal invasion. Moreover, protection was restored by treatment with bradykinin, the major end-product of KKS activation, which was mediated dominantly via bradykinin receptor b1. These data show that IL-17R signaling in RTEC is necessary and likely sufficient for IL-17-mediated renal defense against fatal systemic C. albicans infection.


Asunto(s)
Lesión Renal Aguda/inmunología , Candidemia/inmunología , Membrana Basal Glomerular/metabolismo , Receptores de Interleucina-17/inmunología , Receptores de Interleucina-17/metabolismo , Transducción de Señal/inmunología , Lesión Renal Aguda/microbiología , Traslado Adoptivo , Animales , Bradiquinina/farmacología , Candida albicans , Células Epiteliales/metabolismo , Femenino , Predisposición Genética a la Enfermedad , Membrana Basal Glomerular/citología , Sistema Calicreína-Quinina/efectos de los fármacos , Sistema Calicreína-Quinina/fisiología , Túbulos Renales/metabolismo , Masculino , Ratones , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Receptores de Interleucina-17/genética , Linfocitos T/inmunología , Linfocitos T/metabolismo
20.
BMC Cancer ; 18(1): 382, 2018 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-29618333

RESUMEN

BACKGROUND: The Kallikrein-Kinin System (KKS) has been found to play a role in tumor progression in several cancers. The KKS metabolic cascade depends on signalling through two cross talking receptors; bradykinin receptor 1 (B1R) and bradykinin receptor 2 (B2R). Activation of the Kinin receptor is responsible for multiple pathophysiologic functions including increase of vascular permeability and induction of host inflammatory responses that exert diverse effects on tumor growth. METHODS: B1R and B2R expression on mouse and human CRC cell lines was investigated. Changes in tumor growth and progression was assessed in male CBA mice bearing colorectal liver metastases (CRLM) following treatment with B1R or B2R blockers. In vitro cultures of human SW-480 and mouse colorectal cancer (MoCR) cell lines were examined for changes in their proliferation and migration properties following treatment with B1R or B2R blockers. RESULTS: Both colorectal cancer cell lines tested strongly positive for B1R and B2R expression. Inhibition of both receptors retarded tumor growth but only B1R blockade significantly reduced tumor load and increased tumor apoptosis. Blockade of either receptor reduced tumor vascularization in vivo and significantly inhibited proliferation and migration of colorectal cancer cells in vitro. CONCLUSION: Taken together, the present study demonstrated that kinin receptor blockade inhibited tumor growth and reduced its invading properties suggesting that KKS manipulation could be a novel target in colorectal cancer therapy.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Sistema Calicreína-Quinina , Animales , Apoptosis/efectos de los fármacos , Antagonistas de los Receptores de Bradiquinina/farmacología , Movimiento Celular , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Masculino , Ratones , Metástasis de la Neoplasia , Estadificación de Neoplasias , Neovascularización Patológica/genética , Receptor de Bradiquinina B1/metabolismo , Receptor de Bradiquinina B2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA