Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 213
Filtrar
1.
J Biochem Mol Toxicol ; 37(8): e23393, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37409694

RESUMEN

Doxorubicin (DOX), an effective and broad-spectrum anthracycline antibiotic, is widely used in the treatment of numerous malignancies. However, dose-dependent cardiotoxicity limits the clinical application of DOX, and the molecular mechanisms are still unknown. In this study, we used the BK receptor B1/B2 double-knockout (B1B2 -/- ) mice to observe the role of BK receptor in cardiotoxicity induced by DOX and the underlying mechanisms. DOX induced myocardial injury with increased serum levels of AST, CK, and LDH, upregulated tissue expression of bradykinin B1/B2 receptor, FABP4 and iNOS, and downregulated expression of eNOS. However, these altered releases of myocardial enzyme and the expression level of iNOS were significantly prevented in the B1B2-/- mice. We concluded that the activation of both B1 and B2 receptors of BK were involved in the DOX-induced acute myocardial injury, possibly mediated through iNOS signaling pathways.


Asunto(s)
Cardiotoxicidad , Lesiones Cardíacas , Ratones , Animales , Cardiotoxicidad/metabolismo , Receptores de Bradiquinina/metabolismo , Receptores de Bradiquinina/uso terapéutico , Doxorrubicina/toxicidad , Miocardio/metabolismo , Transducción de Señal , Lesiones Cardíacas/metabolismo , Estrés Oxidativo , Apoptosis , Miocitos Cardíacos/metabolismo
2.
J Photochem Photobiol B ; 239: 112648, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36641883

RESUMEN

Cancer molecular imaging using specific probes designed to identify target proteins in cancer is a powerful tool to guide therapeutic selection, patient management, and follow-up. We demonstrated that icatibant may be used as a targeting probe for the significantly upregulated bradykinin B2R in colorectal cancer (CRC). Icatibant-based probes with high affinity towards bradykinin B2R were identified. The near-infrared (NIR) fluorescent dye conjugate MPA-PEG3-k-Icatibant and radioconjugate [99mTc]Tc-HYNIC-PEG4-Icatibant exhibited favourable selective and specific uptake in tumours when the subcutaneous and orthotopic colorectal tumour-bearing mouse models were imaged using NIR fluorescence imaging and Single-Photon Emission Computed Tomography-Computed Tomography (SPECT-CT), respectively. The tracer of [99mTc]Tc-HYNIC-PEG4-Icatibant accumulated in tumours according to biodistribution studies and peaked at 4 h with an uptake value of 3.41 ± 0.27%ID/g in HT29 tumour-bearing nude mice following intravenous injection (i.v.). The tumour-to-colorectal signal ratios were 5.03 ± 0.37, 15.45 ± 0.32, 13.58 ± 1.19 and 11.33 ± 1.73 1, 2, 4 and 6 h after tail-veil injection, respectively. Overall, in the wake of rapid and precise tumour delineation and penetration characteristics, icatibant-based probes represent promising high-contrast molecular imaging probes for the detection of bradykinin B2R.


Asunto(s)
Bradiquinina , Neoplasias Colorrectales , Receptores de Bradiquinina , Tomografía Computarizada de Emisión de Fotón Único , Animales , Ratones , Bradiquinina/metabolismo , Línea Celular Tumoral , Neoplasias Colorrectales/diagnóstico por imagen , Neoplasias Colorrectales/metabolismo , Ligandos , Ratones Desnudos , Distribución Tisular , Tomografía Computarizada de Emisión de Fotón Único/métodos , Tomografía Computarizada por Rayos X , Receptores de Bradiquinina/metabolismo
3.
Int Immunopharmacol ; 110: 108984, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35780642

RESUMEN

The centrally acting antitussive opiate derivative, noscapine, has been claimed to be a non-competitive bradykinin B2 receptor antagonist. Raloxifene, a selective estrogen receptor modulator, was predicted to bind the bradykinin B2 receptor and to exert a partial agonist activity. These intriguing claims suggest that new molecular scaffolds ("chemotypes") may be identified for small molecule ligands of kinin receptors and that some off-target effects of noscapine or raloxifene may be mediated by bradykinin B2 receptors. An established contractile bioassay for ligands of the bradykinin B2 receptor, the isolated human umbilical vein, was exploited to characterize the inhibitory effect of noscapine and raloxifene on the B2 receptor-mediated contractile response to bradykinin. Observed effects were compared with those of the peptide antagonist icatibant, a potent, selective and competitive B2 receptor antagonist. Our results indicate that neither noscapine (2.5 µM) nor raloxifene (20 µM) behave as B2 receptor antagonists in concentrations that vastly exceeded an effective concentration of the control antagonist, icatibant; further, none of these drugs had direct contractile effects. It is suggested that the previously reported B2 receptor inhibitory effect of noscapine, a putative sigma-receptor agonist, might result from an indirect physiological antagonism, while raloxifene did not appear to have any significant affinity for the B2 receptors.


Asunto(s)
Noscapina , Receptores de Bradiquinina , Bioensayo , Bradiquinina/metabolismo , Antagonistas de los Receptores de Bradiquinina , Humanos , Noscapina/farmacología , Clorhidrato de Raloxifeno/farmacología , Receptor de Bradiquinina B1 , Receptor de Bradiquinina B2 , Receptores de Bradiquinina/metabolismo , Venas Umbilicales/metabolismo
4.
PLoS One ; 17(5): e0267845, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35617279

RESUMEN

The kallikrein-kinin system has been implicated in body weight and glucose homeostasis. Their major effectors act by binding to the kinin B2 and B1 receptors. It was assessed the role of the kinin B1 receptor in weight and glucose homeostasis in B1 receptor knockout mice (B1RKO) subjected to a cafeteria diet (CAF). Wild-type (WT) and B1RKO male mice (C57BL/6 background; 8 weeks old) were fed a standard diet (SD) or CAF for 14 weeks, ad libitum, and four groups were formed: WT-SD; B1RKO-SD; WT-CAF; B1RKO-CAF. Body weight and food intake were assessed weekly. It was performed glucose tolerance (GTT) and insulin tolerance tests (ITT), and HOMA-IR, HOMA-ß and HOMA-ß* 1/HOMA-IR were calculated. Islets from WT and B1RKO were isolated in order to measure the insulin secretion. Western blot was used to assess the hepatic AKT phosphorylation and qPCR to assess gene expression. CAF induced a higher body mass gain in B1RKO compared to WT mice. CAF diet increased epididymal fat depot mass, hepatic fat infiltration and hepatic AKT phosphorylation in both genotypes. However, B1RKO mice presented lower glycemic response during GTT when fed with CAF, and a lower glucose decrease in the ITT. This higher resistance was overcomed with higher insulin secretion when stimulated by high glucose, resulting in higher glucose uptake in the GTT when submitted to CAF, despite lower insulin sensitivity. Islets from B1RKO delivered 4 times more insulin in 3-month-old mice than islets from WT. The higher insulin disposition index and high insulin delivery of B1RKO can explain the decreased glucose excursion during GTT. In conclusion, CAF increased the ß-cell function in B1RKO mice, compensated by the diet-induced insulin resistance and resulting in a healthier glycemic response despite the higher weight gain.


Asunto(s)
Hiperinsulinismo , Resistencia a la Insulina , Receptores de Bradiquinina/metabolismo , Animales , Glucemia/metabolismo , Dieta , Dieta Alta en Grasa , Glucosa/metabolismo , Homeostasis , Insulina/metabolismo , Resistencia a la Insulina/fisiología , Cininas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas c-akt , Aumento de Peso
5.
Int Immunopharmacol ; 105: 108547, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35066448

RESUMEN

BACKGROUND: Tissue kallikrein offers a wide spectrum of biological activity in the protection against various types of injury. However, information on its role in tacrolimus (TAC)-induced renal injury is limited. OBJECTIVES: This study aimed to assess the beneficial effects of pancreatic kininogenase (PK) in a rat model of chronic TAC nephrotoxicity and in vitro. METHODS: Sprague Dawley rats were treated daily with either TAC or PK or a combination of the two for four weeks. The influence of PK on renal injury was examined in terms of renal function, histopathology, cytokine expression, oxidative stress, intracellular organelles, programmed cell death, and PI3K/AKT signaling. Human kidney proximal tubular (HK-2) cells and mouse mesangial (SV40 MES13) cells treated with TAC and PK were also studied. RESULTS: PK treatment improved renal function and histopathology. This effect was paralleled by downregulation of proinflammatory and profibrotic cytokine expression. TAC-induced oxidative stress was closely associated with endoplasmic reticulum stress and mitochondrial dysfunction, resulting in excessive programmed cell death (apoptosis and autophagy) that was significantly abrogated by concurrent PK interference with PI3K/AKT signaling. PK also stimulated bradykinin receptor 1 (B1R) and B2R mRNA synthesis and increased bioactive nitric oxide (NO) and cAMP concentrations in TAC-treated kidneys. Blockade of either B1R or B2R eliminated the renoprotective effects of PK. In HK-2 and SV40 MES13 cells, PK decreased TAC-induced overproduction of intracellular reactive oxygen species and inhibited apoptotic cells, whereas cell viability was improved. Moreover, activated PI3K/AKT signaling in HK-2 cells was inhibited by PK and the PI3K inhibitor, LY294002. CONCLUSIONS: These findings indicate that PK treatment protects against chronic TAC nephrotoxicity via inhibition of PI3K/AKT signaling.


Asunto(s)
Fosfatidilinositol 3-Quinasas , Tacrolimus , Animales , Apoptosis , Riñón , Ratones , Estrés Oxidativo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Bradiquinina/metabolismo , Tacrolimus/farmacología , Calicreínas de Tejido/metabolismo , Calicreínas de Tejido/farmacología
6.
J Pharm Pharmacol ; 73(12): 1643-1651, 2021 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-34061162

RESUMEN

OBJECTIVES: Neuropathic pain (NP) represents a broad scope of various pathological ramifications of the nervous system. Remimazolam is a proved sedative in treating neuropathic pain. Considering the Bradykinin receptor's vital role and the potentials of Bradykinin receptor B1 (BDKRB1) in the neuropathic pain-signalling pathway, we nominated them as a primary target for remimazolam. METHODS: In this study, rats were injected with complete freund's adjuvant (CFA) to construct NP models in vivo. BV2 microglia cells were treated with LPS to establish NP model in vitro. qRT-PCR, ELISA, western blot and immunofluorescence were applied to determine gene expression. KEY FINDINGS: Our findings revealed that BDKRB1 was overexpressed in NP models in vivo, while R715 (an antagonist of BDKRB1) suppressed the levels of BDKRB1 and inhibited the hyperpathia induced by spinal nerve litigation surgery. Moreover, remimazolam inactivated BDKRB1 signalling via suppressing NF-κB translocation and decreased the release of pro-inflammatory cytokines. Additionally, remimazolam suppressed the translocation of NF-κB, and inhibited autophagic lysosome formation in vivo and in vitro. However, R838 (an agonist of BDKRB1) reversed the effects of remimazolam. CONCLUSIONS: Remimazolam downregulated BDKRB1, inhibited BDKRB1/RAS/MEK signalling pathway and regulated the autophagic lysosome induction, exhibiting a better outcome in the NP.


Asunto(s)
Analgésicos/farmacología , Benzodiazepinas/farmacología , Microglía/efectos de los fármacos , Neuralgia/metabolismo , Receptores de Bradiquinina/metabolismo , Analgésicos/uso terapéutico , Animales , Autofagia , Benzodiazepinas/uso terapéutico , Línea Celular , Citocinas/metabolismo , Modelos Animales de Enfermedad , Adyuvante de Freund , Hiperalgesia , Lisosomas/metabolismo , Ratones , FN-kappa B/metabolismo , Neuralgia/tratamiento farmacológico , Ratas Sprague-Dawley , Transducción de Señal
7.
Physiol Rep ; 9(9): e14863, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33991464

RESUMEN

Intermittent hypoxia (IH) is a feature of obstructive sleep apnea (OSA), a condition highly associated with hypertension-related cardiovascular diseases. Repeated episodes of IH contribute to imbalance of angiogenic growth factors in the hypertrophic heart, which is key in the progression of cardiovascular complications. In particular, the interaction between vascular endothelial growth factor (VEGF) and the kallikrein-kinin system (KKS) is essential for promoting angiogenesis. However, researchers have yet to investigate experimental models of IH that reproduce OSA, myocardial angiogenesis, and expression of KKS components. We examined temporal changes in cardiac angiogenesis in a mouse IH model. Adult male C57BI/6 J mice were implanted with Matrigel plugs and subjected to IH for 1-5 weeks with subsequent weekly histological evaluation of vascularization. Expression of VEGF and KKS components was also evaluated. After 3 weeks, in vivo myocardial angiogenesis and capillary density were decreased, accompanied by a late increase of VEGF and its type 2 receptor. Furthermore, IH increased left ventricular myocardium expression of the B2 bradykinin receptor, while reducing mRNA levels of B1 receptor. These results suggest that in IH, an unexpected response of the VEGF and KKS systems could explain the reduced capillary density and impaired angiogenesis in the hypoxic heart, with potential implications in hypertrophic heart malfunction.


Asunto(s)
Cardiomegalia/metabolismo , Hipoxia/metabolismo , Cininas/metabolismo , Miocardio/metabolismo , Neovascularización Fisiológica , Apnea Obstructiva del Sueño/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Capilares/metabolismo , Capilares/fisiología , Cardiomegalia/complicaciones , Vasos Coronarios/metabolismo , Vasos Coronarios/fisiología , Hipoxia/complicaciones , Calicreínas/genética , Calicreínas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Bradiquinina/genética , Receptores de Bradiquinina/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/genética , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Apnea Obstructiva del Sueño/complicaciones , Factor A de Crecimiento Endotelial Vascular/genética
8.
Eur J Pharmacol ; 895: 173870, 2021 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-33476653

RESUMEN

Fibromyalgia is a potentially disabling chronic disease, characterized by widespread pain and a range of comorbidities such as hypertension. Among the mechanisms involved in fibromyalgia-like pain symptoms are kinins and their B1 and B2 receptors. Moreover, angiotensin I converting enzyme (ACE) inhibitors, commonly used as antihypertensive drugs, can enhance pain by blocking the degradation of peptides such as substance P and bradykinin, besides enhancing kinin receptors signalling. We investigated the effect of ACE inhibitors on reserpine-induced fibromyalgia-like pain symptoms and the involvement of kinins in this effect in mice. Nociceptive parameters (mechanical and cold allodynia and overt nociception) were evaluated after ACE inhibitors administration in mice previously treated with reserpine. The role of kinin B1 and B2 receptors was investigated using pharmacological antagonism. Additionally, bradykinin levels, as well as the activity of ACE and kininase I, were measured in the sciatic nerve, spinal cord and cerebral cortex of the mice. The ACE inhibitors enalapril and captopril enhanced reserpine-induced mechanical allodynia, and this increase was prevented by kinin B1 and B2 receptor antagonists. Substance P and bradykinin caused overt nociception and increased mechanical allodynia in animals treated with reserpine. Reserpine plus ACE inhibitors increased bradykinin-related peptide levels and inhibited ACE activity in pain modulation structures. Since hypertension is a frequent comorbidity affecting fibromyalgia patients, hypertension treatment with ACE inhibitors in these patients should be reviewed once this could enhance fibromyalgia-like pain symptoms. Thus, the treatment of hypertensive patients with fibromyalgia could include other classes of antihypertensive drugs, different from ACE inhibitors.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/toxicidad , Fibromialgia/inducido químicamente , Sistema Nervioso/efectos de los fármacos , Dolor Nociceptivo/inducido químicamente , Umbral del Dolor/efectos de los fármacos , Peptidil-Dipeptidasa A/metabolismo , Receptores de Bradiquinina/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Captopril/toxicidad , Modelos Animales de Enfermedad , Enalapril/toxicidad , Fibromialgia/enzimología , Fibromialgia/fisiopatología , Masculino , Ratones , Sistema Nervioso/enzimología , Sistema Nervioso/fisiopatología , Dolor Nociceptivo/enzimología , Dolor Nociceptivo/fisiopatología , Reserpina , Transducción de Señal
9.
Sci Rep ; 9(1): 2973, 2019 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-30814538

RESUMEN

The regulation of the kallikrein-kinin system is an important mechanism controlling vasodilation and promoting inflammation. We aimed to investigate the role of Toll-like receptor 2 (TLR2) in regulating kinin B1 and B2 receptor expression in human gingival fibroblasts and in mouse gingiva. Both P. gingivalis LPS and the synthetic TLR2 agonist Pam2CSK4 increased kinin receptor transcripts. Silencing of TLR2, but not of TLR4, inhibited the induction of kinin receptor transcripts by both P. gingivalis LPS and Pam2CSK4. Human gingival fibroblasts (HGF) exposed to Pam2CSK4 increased binding sites for bradykinin (BK, B2 receptor agonist) and des-Arg10-Lys-bradykinin (DALBK, B1 receptor agonist). Pre-treatment of HGF for 24 h with Pam2CSK4 resulted in increased PGE2 release in response to BK and DALBK. The increase of B1 and B2 receptor transcripts by P. gingivalis LPS was not blocked by IL-1ß neutralizing antibody; TNF-α blocking antibody did not affect B1 receptor up-regulation, but partially blocked increase of B2 receptor mRNA. Injection of P. gingivalis LPS in mouse gingiva induced an increase of B1 and B2 receptor mRNA. These data show that activation of TLR2 in human gingival fibroblasts as well as in mouse gingival tissue leads to increase of B1 and B2 receptor mRNA and protein.


Asunto(s)
Receptores de Bradiquinina/genética , Receptor Toll-Like 2/metabolismo , Adulto , Animales , Bradiquinina/metabolismo , Femenino , Fibroblastos/metabolismo , Encía/metabolismo , Humanos , Inflamación/metabolismo , Cininas/metabolismo , Lipopéptidos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Receptor de Bradiquinina B1/genética , Receptor de Bradiquinina B2/genética , Receptores de Bradiquinina/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
10.
Prog Neurobiol ; 165-167: 26-50, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29355711

RESUMEN

Neurological disorders represent major health concerns in terms of comorbidity and mortality worldwide. Despite a tremendous increase in our understanding of the pathophysiological processes involved in disease progression and prevention, the accumulated knowledge so far resulted in relatively moderate translational benefits in terms of therapeutic interventions and enhanced clinical outcomes. Aiming at specific neural molecular pathways, different strategies have been geared to target the development and progression of such disorders. The kallikrein-kinin system (KKS) is among the most delineated candidate systems due to its ubiquitous roles mediating several of the pathophysiological features of these neurological disorders as well as being implicated in regulating various brain functions. Several experimental KKS models revealed that the inhibition or stimulation of the two receptors of the KKS system (B1R and B2R) can exhibit neuroprotective and/or adverse pathological outcomes. This updated review provides background details of the KKS components and their functions in different neurological disorders including temporal lobe epilepsy, traumatic brain injury, stroke, spinal cord injury, Alzheimer's disease, multiple sclerosis and glioma. Finally, this work will highlight the putative roles of the KKS components as potential neurotherapeutic targets and provide future perspectives on the possibility of translating these findings into potential clinical biomarkers in neurological disease.


Asunto(s)
Biomarcadores/metabolismo , Sistema Calicreína-Quinina/fisiología , Cininas/metabolismo , Enfermedades del Sistema Nervioso/metabolismo , Receptores de Bradiquinina/metabolismo , Transducción de Señal/fisiología , Animales , Humanos
11.
Inflamm Res ; 67(4): 301-314, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29260240

RESUMEN

OBJECTIVE: We investigated whether: (1) P2 × 7 receptor activation by its agonist (BzATP) induces articular hyperalgesia in the rat's knee joint via inflammatory mechanisms and (2) activation of P2 × 7 receptors by endogenous ATP contributes to the articular hyperalgesia induced by bradykinin, TNF-α, IL-1ß, CINC-1, PGE2, and dopamine. METHODS: The articular hyperalgesia was quantified using the rat knee joint incapacitation test. The knee joint inflammation, characterized by the concentration of pro-inflammatory cytokines and by neutrophil migration, was quantified in the synovial lavage fluid by ELISA and myeloperoxidase enzyme activity assay, respectively. RESULTS: BzATP induced a dose-dependent articular hyperalgesia in the rat's knee joint that was significantly reduced by the selective antagonists for P2 × 7, bradykinin B1 or B2 receptors, ß1 or ß2 adrenoceptors, and by pre-treatment with Indomethacin. BzATP induced a local increase of TNF-α, IL-1ß, IL-6, and CINC-1 concentration and neutrophil migration into the knee joint. The co-administration of the selective P2 × 7 receptor antagonist A-740003 significantly reduced the articular hyperalgesia induced by bradykinin and dopamine, but not by TNF-α, IL-1ß, CINC-1, and PGE2. CONCLUSIONS: P2 × 7 receptor activation induces articular hyperalgesia mediated by the previous inflammatory mediator release. P2 × 7 receptor-induced articular hyperalgesia is sustained by the involvement of this purinergic receptor in bradykinin and dopamine-induced hyperalgesia in the knee joint.


Asunto(s)
Hiperalgesia/etiología , Articulación de la Rodilla/metabolismo , Receptores de Bradiquinina/metabolismo , Receptores Dopaminérgicos/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Adenosina Trifosfato/análogos & derivados , Antagonistas Adrenérgicos beta/farmacología , Animales , Bradiquinina , Antagonistas de los Receptores de Bradiquinina/farmacología , Citocinas/metabolismo , Dopamina , Hiperalgesia/inducido químicamente , Hiperalgesia/metabolismo , Mediadores de Inflamación/metabolismo , Masculino , Antagonistas de Prostaglandina/farmacología , Antagonistas del Receptor Purinérgico P2/farmacología , Agonistas del Receptor Purinérgico P2X/farmacología , Antagonistas del Receptor Purinérgico P2X/farmacología , Ratas Wistar
12.
Wiad Lek ; 71(8): 1615-1620, 2018.
Artículo en Polaco | MEDLINE | ID: mdl-30684349

RESUMEN

Kallikreins cleave kininogens to release kinins. Kinins exert their biological effect by activating constitutive bradykinin receptor-2 (BR2) and inducible by inflammatory cytokines bradykinin receptor-1 (BR1). Studies in animal models and some clinical observations indicate tat the activation of kallikrein - kinin system may have relevance to central nervous system (CNS) diseases, including multiple sclerosis, Alzheimer's disease, epilepsy as well as cerebral ischemia and neoplasmatic tumors. The actions of kinins include vasodilatation and increased vascular permeability may contribute to blood-brain barrier disruption. Kinins evoke pain, and stimulate of endothelial cells, white blood cells, astrocytes and microgia cells to release of prostanoids, cytokines, free radicals, nitric oxide. Kinins stimulate angiogenesis and proliferation of tumor cells. These events lead to neural tissue damage and long lasting disturbances in blood-brain barrier function. In animal models the overexpression of genes and proteins of tissue kallikrens, kininogen as well as RB1 and RB2 has been observed. Kinin receptors antagonists, especially B1R blockade decreased morphological and biochemical features of CNS inflammation. On the other hand in brain tumor models RB1 and RB2 activation has been shown to mediate reversible blood-brain barrier permeability to enhance anti-cancer drug delivery, which may have therapeutic potential.


Asunto(s)
Enfermedades del Sistema Nervioso Central/fisiopatología , Sistema Calicreína-Quinina , Animales , Antagonistas de los Receptores de Bradiquinina/uso terapéutico , Humanos , Inflamación , Calicreínas/metabolismo , Cininas/metabolismo , Receptores de Bradiquinina/metabolismo
13.
Eur J Pain ; 22(3): 501-510, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29034546

RESUMEN

BACKGROUND: Endometriosis is a gynaecological disease exhibiting severe pelvic pain, but the mechanism of pain production remains unknown. Bradykinin (BK) is known as an inflammatory mediator, and shows elevated levels in inflammatory diseases such as rheumatoid arthritis. In the present study, we evaluated whether BK is involved in endometriosis-related pain. METHODS: Endometriotic lesions were used for immunohistochemistry. Primary cultures of endometriotic stromal cells (ESC) were stimulated with IL-1ß and/or BK. Quantitative RT-PCR was used to evaluate the mRNA expressions of BK receptors (BKR) and endothelin-1 in ESC. The concentration of endothelin-1 in cystic fluid of endometrioma or non-endometrioma was measured with ELISA. The conditioned medium of ESC stimulated with IL-1ß and/or BK was injected intraplantarly in mice, and evaluated whether pain-related licking behaviour was elicited. RESULTS: The expressions of BK and BKR in endometriotic lesions were observed by immunohistochemistry. In vitro experiments showed that IL-1ß induced BKR-B1 and B2 on ESC. Activation of these receptors by BK significantly induced endothelin-1 expression in ESC, which was negated completely by HOE-140, a BKR-B2 antagonist. The cystic fluid of endometrioma contained higher amount of endothelin-1 compared to non-endometrioma. Intraplantar injection of the conditioned medium of ESC treated with IL-1ß and BK significantly induced licking behaviour, which was suppressed with BQ-123, an endothelin type-A receptor antagonist. CONCLUSIONS: The present study demonstrated the presence and the function of the BK axis in endometriosis, and established a potential new therapy target for endometriosis-related pain. SIGNIFICANCE: The present study demonstrated (1) the presence and the function of the BK system in endometriosis, (2) activation of BKR induced endothelin-1 in endometriotic lesion and (3) blocking endothelin-1 was effective to decrease pain.


Asunto(s)
Bradiquinina/metabolismo , Endometriosis/metabolismo , Endotelina-1/metabolismo , Dolor/metabolismo , Receptores de Bradiquinina/metabolismo , Células del Estroma/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Bradiquinina/análogos & derivados , Bradiquinina/farmacología , Antagonistas del Receptor de Bradiquinina B2/farmacología , Estudios de Casos y Controles , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Líquido Quístico/metabolismo , Endotelina-1/efectos de los fármacos , Endotelina-1/genética , Endotelina-1/farmacología , Femenino , Humanos , Interleucina-1beta/farmacología , Ratones , Enfermedades del Ovario/metabolismo , Enfermedades Peritoneales/metabolismo , ARN Mensajero/metabolismo , Receptores de Bradiquinina/efectos de los fármacos , Receptores de Bradiquinina/genética , Células del Estroma/efectos de los fármacos
14.
Peptides ; 90: 10-16, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28192151

RESUMEN

Angiotensin-(1-7) [Ang-(1-7)] exhibits blood pressure lowering actions, inhibits cell growth, and reduces tissue inflammation and fibrosis which may functionally antagonize an activated Ang II-AT1 receptor axis. Since the vascular actions of Ang-(1-7) and the associated receptor/signaling pathways vary in different vascular beds, the current study established the vasorelaxant properties of the heptapeptide in the renal artery of male Wistar male rats. Ang-(1-7) produced an endothelium-dependent vasodilator relaxation of isolated renal artery segments pre-contracted by a sub-maximal concentration of phenylephrine (PE) (3×10-7M). Ang-(1-7) induced vasodilation of the rat renal artery with an ED50 of 3±1nM and a maximal response of 42±5% (N=10). The two antagonists (10-5M each) for the AT7/Mas receptor (MasR) [D-Pro7]-Ang-(1-7) and [D-Ala7]-Ang-(1-7) significantly reduced the maximal response to 12±1% and 18±3%, respectively. Surprisingly, the AT2R receptor antagonist PD123319, the AT1R antagonist losartan and B2R antagonist HOE140 (10-6M each) also significantly reduced Ang-(1-7)-induced relaxation to 12±2%, 22±3% and 14±7%, respectively. Removal of the endothelium or addition of the soluble guanylate cyclase (sGC) inhibitor ODQ (10-5M) essentially abolished the vasorelaxant response to Ang-(1-7) (10±4% and 10±2%, P <0.05). Finally, the NOS inhibitor LNAME (10-4M) reduced the response to 13±2% (p<0.05), but the cyclooxygenase inhibitor indomethacin failed to block the Ang-(1-7) response. We conclude that Ang-(1-7) exhibits potent vasorelaxant actions in the isolated renal artery that are dependent on an intact endothelium and the apparent stimulation of a NO-sGC pathway. Moreover, Ang-(1-7)-dependent vasorelaxation was sensitive to antagonists against the AT7/Mas, AT1, AT2 and B2 receptor subtypes.


Asunto(s)
Angiotensina I/metabolismo , Fragmentos de Péptidos/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Receptores de Bradiquinina/metabolismo , Arteria Renal/metabolismo , Angiotensina I/administración & dosificación , Antagonistas de Receptores de Angiotensina/administración & dosificación , Animales , Presión Sanguínea/efectos de los fármacos , Antagonistas de los Receptores de Bradiquinina/administración & dosificación , Guanilato Ciclasa/metabolismo , Humanos , Imidazoles/administración & dosificación , Losartán/administración & dosificación , NG-Nitroarginina Metil Éster/administración & dosificación , Óxido Nítrico/metabolismo , Fragmentos de Péptidos/administración & dosificación , Fenilefrina/administración & dosificación , Proto-Oncogenes Mas , Piridinas/administración & dosificación , Ratas , Arteria Renal/efectos de los fármacos , Arteria Renal/patología , Transducción de Señal/efectos de los fármacos , Vasodilatación/genética
15.
Eur J Oral Sci ; 125(1): 18-27, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28032657

RESUMEN

Bradykinin is an important peptide modulator that affects the function of neurons and immune cells. However, there is no evidence of the bradykinin receptors and their functions in human salivary glands. Here we have identified and characterized bradykinin receptors on human submandibular gland cells. Both bradykinin B1 and B2 receptors are expressed on human submandibular gland cells, A253 cells, and HSG cells. Bradykinin increased the intracellular Ca2+ concentration ([Ca2+ ]i ) in a concentration-dependent manner. Interestingly, a specific agonist of the B1 receptor did not have any effect on [Ca2+ ]i in HSG cells, whereas specific agonists of the B2 receptor had a Ca2+ mobilizing effect. Furthermore, application of the B1 receptor antagonist, R715, did not alter the bradykinin-mediated increase in cytosolic Ca2+ , whereas the B2 receptor antagonist, HOE140, showed a strong inhibitory effect, which implies that bradykinin B2 receptors are functional in modulating the concentration of cytosolic Ca2+ . Bradykinin did not affect a carbachol-induced rise of [Ca2+ ]i and did not modulate translocation of aquaporin-5. However, bradykinin did promote the expression of proinflammatory cytokines, including tumor necrosis factor-α (TNF-α), implying the role of bradykinin in salivary gland inflammation. These data suggest that bradykinin receptors are involved in Ca2+ signaling in human submandibular gland cells and serve a unique role, which is separate from that of other salivary gland G protein-coupled receptors.


Asunto(s)
Citocinas/metabolismo , Receptores de Bradiquinina/metabolismo , Glándulas Salivales/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Acuaporina 5/metabolismo , Western Blotting , Bradiquinina/análogos & derivados , Bradiquinina/farmacología , Calcio/metabolismo , Carbacol/farmacología , Línea Celular , Células Cultivadas , AMP Cíclico/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Masculino , Persona de Mediana Edad , Reacción en Cadena en Tiempo Real de la Polimerasa , Glándulas Salivales/citología , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
16.
Mol Neurobiol ; 54(10): 7824-7837, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27844290

RESUMEN

Paclitaxel is a chemotherapeutic agent used to treat solid tumours. However, it causes an acute and neuropathic pain syndrome that limits its use. Among the mechanisms involved in neuropathic pain caused by paclitaxel is activation of kinin receptors. Angiotensin converting enzyme (ACE) inhibitors can enhance kinin receptor signalling. The goal of this study was to evaluate the role of kinins on paclitaxel-associated acute pain syndromes (P-APS) and the effect of ACE inhibition on P-APS and paclitaxel-associated chronic peripheral neuropathy (P-CPN) in mice. Herein, we show that paclitaxel caused mechanical allodynia and spontaneous nociceptive behaviour that was reduced by antagonists of kinin receptors B1 (DALBk and SSR240612) and B2 (Hoe140 and FR173657). Moreover, enalapril (an ACE inhibitor) enhanced the mechanical allodynia induced by a low dose of paclitaxel. Likewise, paclitaxel injection inhibited ACE activity and increased the expressions of B1 and B2 receptors and bradykinin-related peptides levels in peripheral tissue. Together, our data support the involvement of kinin receptors in the P-APS and suggest kinin receptor antagonists to treat this syndrome. Because hypertension is the most frequent comorbidity affecting cancer patients, treatment of hypertension with ACE inhibitors in patients undergoing paclitaxel chemotherapy should be reviewed, since this could enhance the P-APS and P-CPN.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/toxicidad , Bradiquinina/metabolismo , Neuralgia/inducido químicamente , Neuralgia/metabolismo , Paclitaxel/toxicidad , Receptores de Bradiquinina/metabolismo , Animales , Antineoplásicos/toxicidad , Sinergismo Farmacológico , Masculino , Ratones , Dimensión del Dolor/efectos de los fármacos , Dimensión del Dolor/métodos
17.
Lasers Med Sci ; 32(1): 87-94, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27726041

RESUMEN

The objective of this study was to evaluate the effects of photobiomodulation therapy (PBMT) on inflammatory indicators, i.e., inflammatory mediators (TNF-α and CINC-1), and pain characterized by hyperalgesia and B1 and B2 receptor activation at 6, 24, and 48 h after papain-induced osteoarthritis (OA) in rats. Fifty-four rats were subjected to hyperalgesia evaluations and then divided randomly into three groups-a control group and two groups OA and OA PBMT group by using laser parameters at wavelength (808 nm), output power (50 mW), energy per point (4 Joules), power density (1.78 W/cm2), laser beam (0.028 cm2), and energy density (144 J/cm2)-the induction of osteoarthritis was then performed with 20-µl injections of a 4 % papain solution dissolved in 10 µl of saline solution, to which 10 µl of cysteine solution (0.03 M). The statistical analysis was performed using two-way ANOVA with Bonferroni's post hoc test for comparisons between the 6, 24, and 48 h and team points within each group, and between the control, injury, and PBMT groups, and p < 0.05 was considered to indicate a significant difference. The hyperalgesia was evaluated at 6, 24, and 48 h after the injury. PBMT at a wavelength of 808 nm and doses of 4 J, administered afterward, promotes increase at the threshold of pressure stimulus at 6, 24, and 48 h after application and promote cytokine attenuation levels (TNF and CINC-1) and bradykinin receptor (B1 and B2) along the experimental period. We conclude that photobiomodulation therapy was able to promote the reduction of proinflammatory cytokines such as TNF-α and CINC-1, to reduce the gene and protein expression of the bradykinin receptor (B1 and B2), as well as increasing the stimulus response threshold of pressure in an experimental model of acute osteoarthritis.


Asunto(s)
Mediadores de Inflamación/metabolismo , Terapia por Luz de Baja Intensidad , Osteoartritis/metabolismo , Osteoartritis/radioterapia , Receptores de Bradiquinina/metabolismo , Enfermedad Aguda , Animales , Quimiocina CXCL1 , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Extremidades/patología , Regulación de la Expresión Génica , Hiperalgesia/complicaciones , Hiperalgesia/genética , Hiperalgesia/metabolismo , Hiperalgesia/patología , Masculino , Osteoartritis/complicaciones , Osteoartritis/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Necrosis Tumoral alfa/metabolismo
18.
Kidney Int ; 90(5): 1023-1036, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27546607

RESUMEN

The kallikrein-kinin system has been shown to be involved in the development of diabetic nephropathy, but specific mechanisms are not fully understood. Here, we determined the renal-protective role of exogenous pancreatic kallikrein in diabetic mice and studied potential mechanisms in db/db type 2 diabetic and streptozotocin-induced type 1 diabetic mice. After the onset of diabetes, mice were treated with either pancreatic kallikrein (db/db+kallikrein, streptozotocin+kallikrein) or saline (db/db+saline, streptozotocin+saline) for 16 weeks, while another group of streptozotocin-induced diabetic mice received the same treatment after onset of albuminuria (streptozotocin'+kallikrein, streptozotocin'+saline). Db/m littermates or wild type mice were used as non-diabetic controls. Pancreatic kallikrein had no effects on body weight, blood glucose and blood pressure, but significantly reduced albuminuria among all three groups. Pathological analysis showed that exogenous kallikrein decreased the thickness of the glomerular basement membrane, protected against the effacement of foot process, the loss of endothelial fenestrae, and prevented the loss of podocytes in diabetic mice. Renal fibrosis, inflammation and oxidative stress were reduced in kallikrein-treated mice compared to diabetic controls. The expression of kininogen1, tissue kallikrein, kinin B1 and B2 receptors were all increased in the kallikrein-treated compared to saline-treated mice. Thus, exogenous pancreatic kallikrein both prevented and ameliorated diabetic nephropathy, which may be mediated by activating the kallikrein-kinin system.


Asunto(s)
Albuminuria/tratamiento farmacológico , Coagulantes/uso terapéutico , Nefropatías Diabéticas/prevención & control , Calicreínas/uso terapéutico , Riñón/efectos de los fármacos , Albuminuria/etiología , Animales , Coagulantes/farmacología , Creatinina/sangre , Nefropatías Diabéticas/complicaciones , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Evaluación Preclínica de Medicamentos , Fibrosis , Inflamación/tratamiento farmacológico , Calicreínas/metabolismo , Calicreínas/farmacología , Riñón/patología , Quininógenos/metabolismo , Masculino , Ratones , Óxido Nítrico/orina , Estrés Oxidativo/efectos de los fármacos , Receptores de Bradiquinina/metabolismo
19.
J Pain ; 17(11): 1183-1197, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27544818

RESUMEN

Chronic vulvar pain is alarmingly common in women of reproductive age and is often accompanied by psychological distress, sexual dysfunction, and a significant reduction in quality of life. Localized provoked vulvodynia (LPV) is associated with intense vulvar pain concentrated in the vulvar vestibule (area surrounding vaginal opening). To date, the origins of vulvodynia are poorly understood, and treatment for LPV manages pain symptoms, but does not resolve the root causes of disease. Until recently, no definitive disease mechanisms had been identified; our work indicates LPV has inflammatory origins, although additional studies are needed to understand LPV pain. Bradykinin signaling is one of the most potent inducers of inflammatory pain and is a candidate contributor to LPV. We report that bradykinin receptors are expressed at elevated levels in LPV patient versus healthy control vestibular fibroblasts, and patient vestibular fibroblasts produce elevated levels of proinflammatory mediators with bradykinin stimulation. Inhibiting expression of one or both bradykinin receptors significantly reduces proinflammatory mediator production. Finally, we determined that bradykinin activates nuclear factor (NF)κB signaling (a major inflammatory pathway), whereas inhibition of NFκB successfully ablates this response. These data suggest that therapeutic agents targeting bradykinin sensing and/or NFκB may represent new, more specific options for LPV therapy. PERSPECTIVE: There is an unmet need for the development of more effective vulvodynia therapies. As we explore the mechanisms by which human vulvar fibroblasts respond to proinflammatory/propain stimuli, we move closer to understanding the origins of chronic vulvar pain and identifying new therapeutic targets, knowledge that could significantly improve patient care.


Asunto(s)
Bradiquinina/metabolismo , Dolor Pélvico/metabolismo , Transducción de Señal/fisiología , Adulto , Bradiquinina/análogos & derivados , Bradiquinina/genética , Bradiquinina/farmacología , Antagonistas de los Receptores de Bradiquinina/farmacología , Estudios de Casos y Controles , Células Cultivadas , Dolor Crónico , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Interleucina-6/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Dolor Pélvico/tratamiento farmacológico , Dolor Pélvico/patología , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptores de Bradiquinina/genética , Receptores de Bradiquinina/metabolismo
20.
Eksp Klin Farmakol ; 79(6): 36-44, 2016.
Artículo en Ruso | MEDLINE | ID: mdl-29782753

RESUMEN

It is known that agonists of adenosine, opioid, and bradykinin receptors mimic the phenomenon of ischemic postconditioning. There is no commonly accepted notion of what adenosine receptor subtypes must be activated to increase cardiac resistance to reperfusion injury. Intravenous infusion of adenosine or intracoronary administration of adenosine produce infarct-limiting effect and contribute to a more complete restoration of coronary blood flow after recanalization of the infarct-related coronary artery. It was confirmed that opioids mimic the phenomenon of postconditioning. According to obtained data, the most promising compounds for the prevention of reperfusion injury of the heart are κ(1)- and δ(2)-opioid receptor agonists, as they produce the infarct-limiting effect, while not reducing the arterial pressure.


Asunto(s)
Circulación Coronaria/efectos de los fármacos , Vasos Coronarios/metabolismo , Poscondicionamiento Isquémico , Miocardio/metabolismo , Agonistas del Receptor Purinérgico P1/uso terapéutico , Receptores de Bradiquinina/agonistas , Receptores Opioides/agonistas , Animales , Vasos Coronarios/patología , Humanos , Miocardio/patología , Receptores de Bradiquinina/metabolismo , Receptores Opioides/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA