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











Base de dados
Intervalo de ano de publicação
1.
Pharmacol Res ; 205: 107242, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38823470

RESUMO

Targeting the CCL2/CCR2 chemokine axis has been shown to be effective at relieving pain in rodent models of inflammatory and neuropathic pain, therefore representing a promising avenue for the development of non-opioid analgesics. However, clinical trials targeting this receptor for inflammatory conditions and painful neuropathies have failed to meet expectations and have all been discontinued due to lack of efficacy. To overcome the poor selectivity of CCR2 chemokine receptor antagonists, we generated and characterized the function of intracellular cell-penetrating allosteric modulators targeting CCR2, namely pepducins. In vivo, chronic intrathecal administration of the CCR2-selective pepducin PP101 was effective in alleviating neuropathic and bone cancer pain. In the setting of bone metastases, we found that T cells infiltrate dorsal root ganglia (DRG) and induce long-lasting pain hypersensitivity. By acting on CCR2-expressing DRG neurons, PP101 attenuated the altered phenotype of sensory neurons as well as the neuroinflammatory milieu of DRGs, and reduced bone cancer pain by blocking CD4+ and CD8+ T cell infiltration. Notably, PP101 demonstrated its efficacy in targeting the neuropathic component of bone cancer pain, as evidenced by its anti-nociceptive effects in a model of chronic constriction injury of the sciatic nerve. Importantly, PP101-induced reduction of CCR2 signaling in DRGs did not result in deleterious tumor progression or adverse behavioral effects. Thus, targeting neuroimmune crosstalk through allosteric inhibition of CCR2 could represent an effective and safe avenue for the management of chronic pain.


Assuntos
Dor Crônica , Gânglios Espinais , Neuralgia , Receptores CCR2 , Animais , Receptores CCR2/antagonistas & inibidores , Receptores CCR2/metabolismo , Dor Crônica/tratamento farmacológico , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Neuralgia/tratamento farmacológico , Neuralgia/metabolismo , Humanos , Dor do Câncer/tratamento farmacológico , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/secundário , Analgésicos/farmacologia , Analgésicos/uso terapêutico , Masculino , Camundongos , Feminino , Camundongos Endogâmicos C57BL
2.
ACS Pharmacol Transl Sci ; 6(2): 290-305, 2023 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-36798478

RESUMO

Apelin is an endogenous peptide that is involved in many diseases such as cardiovascular diseases, obesity, and cancer, which has made it an attractive target for drug discovery. Herein, we explore the penultimate and final sequence positions of [Pyr1]-apelin-13 (Ape13) via C-terminal N α-alkylated amide bonds and the introduction of positive charges, potentially targeting the allosteric sodium pocket, by assessing the binding affinity and signaling profiles at the apelin receptor (APJ). Synthetic analogues modified within this segment of Ape13 showed high affinity (K i 0.12-0.17 nM vs Ape13 K i 0.7 nM), potent Gαi1 activation (EC50 Gαi1 0.4-0.9 nM vs Ape13 EC50 1.1 nM), partial agonist behavior disfavoring ß-arrestin 2 recruitment for positively charged ligands (e.g., 49 (SBL-AP-058), EC50 ß-arr2 275 nM, E max 54%) and high plasma stability for N-alkyl ligands (t 1/2 > 7 h vs Ape13 t 1/2 0.5 h). Combining the benefits of the N α-alkylated amide bond with the guanidino substitution in a constrained ligand led to 63 (SBL-AP-049), which displayed increased plasma stability (t 1/2 5.3 h) and strong reduction of ß-arrestin 2 signaling with partial maximal efficacy (EC50 ß-arr 864 nM, E max 48%), significantly reducing the hypotensive effect in vivo.

3.
Cancer Metastasis Rev ; 40(2): 427-445, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33973098

RESUMO

Breast and prostate cancers have a great propensity to metastasize to long bones. The development of bone metastases is life-threatening, incurable, and drastically reduces patients' quality of life. The chemokines CCL2 and CXCL12 and their respective receptors, CCR2 and CXCR4, are central instigators involved in all stages leading to cancer cell dissemination and secondary tumor formation in distant target organs. They orchestrate tumor cell survival, growth and migration, tumor invasion and angiogenesis, and the formation of micrometastases in the bone marrow. The bone niche is of particular importance in metastasis formation, as it expresses high levels of CCL2 and CXCL12, which attract tumor cells and contribute to malignancy. The limited number of available effective treatment strategies highlights the need to better understand the pathophysiology of bone metastases and reduce the skeletal tumor burden in patients diagnosed with metastatic bone disease. This review focuses on the involvement of the CCL2/CCR2 and CXCL12/CXCR4 chemokine axes in the formation and development of bone metastases, as well as on therapeutic perspectives aimed at targeting these chemokine-receptor pairs.


Assuntos
Neoplasias Ósseas/imunologia , Neoplasias Ósseas/secundário , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Quimiocina CCL2/imunologia , Quimiocina CXCL12/imunologia , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/patologia , Animais , Feminino , Humanos , Masculino
4.
Neurosci Biobehav Rev ; 125: 168-192, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33582232

RESUMO

Chronic pain is a major global health issue that affects all populations regardless of sex, age, ethnicity/race, or country of origin, leading to persistent physical and emotional distress and to the loss of patients' autonomy and quality of life. Despite tremendous efforts in the elucidation of the mechanisms contributing to the pathogenesis of chronic pain, the identification of new potential pain targets, and the development of novel analgesics, the pharmacological treatment options available for pain management remain limited, and most novel pain medications have failed to achieve advanced clinical development, leaving many patients with unbearable and undermanaged pain. Sex-specific susceptibility to chronic pain conditions as well as sex differences in pain sensitivity, pain tolerance and analgesic efficacy are increasingly recognized in the literature and have thus prompted scientists to seek mechanistic explanations. Hence, recent findings have highlighted that the signaling mechanisms underlying pain hypersensitivity are sexually dimorphic, which sheds light on the importance of conducting preclinical and clinical pain research on both sexes and of developing sex-specific pain medications. This review thus focuses on the clinical and preclinical evidence supporting the existence of sex differences in pain neurobiology. Attention is drawn to the sexually dimorphic role of glial and immune cells, which are both recognized as key players in neuroglial maladaptive plasticity at the origin of the transition from acute pain to chronic pathological pain. Growing evidence notably attributes to microglial cells a pivotal role in the sexually dimorphic pain phenotype and in the sexually dimorphic analgesic efficacy of opioids. This review also summarizes the recent advances in understanding the pathobiology underpinning the development of pain hypersensitivity in both males and females in different types of pain conditions, with particular emphasis on the mechanistic signaling pathways driving sexually dimorphic pain responses.


Assuntos
Dor , Qualidade de Vida , Analgésicos Opioides , Feminino , Humanos , Masculino , Neuroglia , Dor/tratamento farmacológico , Caracteres Sexuais
5.
J Med Chem ; 64(1): 602-615, 2021 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-33350824

RESUMO

ELABELA (ELA) is the second endogenous ligand of the apelin receptor (APJ). Although apelin-13 and ELA both target APJ, there is limited information on structure-activity relationship (SAR) of ELA. In the present work, we identified the shortest bioactive C-terminal fragment ELA23-32, which possesses high affinity for APJ (Ki 4.6 nM) and produces cardiorenal effects in vivo similar to those of ELA. SAR studies on conserved residues (Leu25, His26, Val29, Pro30, Phe31, Pro32) show that ELA and apelin-13 may interact differently with APJ. His26 and Val29 emerge as important for ELA binding. Docking and binding experiments suggest that Phe31 of ELA may bind to a tight groove distinct from that of Phe13 of Ape13, while the Phe13 pocket may be occupied by Pro32 of ELA. Further characterization of signaling profiles on the Gαi1, Gα12, and ß-arrestin2 pathways reveals the importance of aromatic residue at the Phe31 or Pro32 position for receptor activation.


Assuntos
Receptores de Apelina/agonistas , Hormônios Peptídicos/farmacologia , Sequência de Aminoácidos , Animais , Receptores de Apelina/metabolismo , Sítios de Ligação , Pressão Sanguínea/efeitos dos fármacos , Biologia Computacional , Coração/efeitos dos fármacos , Coração/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/química , Ligantes , Masculino , Hormônios Peptídicos/química , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade
6.
Pharmaceuticals (Basel) ; 13(10)2020 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-33003415

RESUMO

The blood-brain barrier (BBB) is a major obstacle to the development of effective diagnostics and therapeutics for brain cancers and other central nervous system diseases. Peptide agonist analogs of kinin B1 and B2 receptors, acting as BBB permeabilizers, have been utilized to overcome this barrier. The purpose of the study was to provide new insights for the potential utility of kinin analogs as brain drug delivery adjuvants. In vivo imaging studies were conducted in various animal models (primary/secondary brain cancers, late radiation-induced brain injury) to quantify BBB permeability in response to kinin agonist administrations. Results showed that kinin B1 (B1R) and B2 receptors (B2R) agonists increase the BBB penetration of chemotherapeutic doxorubicin to glioma sites, with additive effects when applied in combination. B2R agonist also enabled extravasation of high-molecular-weight fluorescent dextrans (155 kDa and 2 MDa) in brains of normal mice. Moreover, a systemic single dose of B2R agonist did not increase the incidence of metastatic brain tumors originating from circulating breast cancer cells. Lastly, B2R agonist promoted the selective delivery of co-injected diagnostic MRI agent Magnevist in irradiated brain areas, depicting increased vascular B2R expression. Altogether, our findings suggest additional evidence for using kinin analogs to facilitate specific access of drugs to the brain.

7.
Eur J Pharmacol ; 848: 80-87, 2019 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-30707956

RESUMO

Opioid and neurotensin (NT) receptors are expressed in both central and peripheral nervous systems where they modulate nociceptive responses. Nowadays, opioid analgesics like morphine remain the most prescribed drugs for the treatment of moderate to severe pain. However, despite their daily used, opioids can produce life-threatening side effects, such as constipation or respiratory depression. Besides, NT analogs exert strong opioid-independent analgesia. Here, we thus hypothesized that the combined use of opioid and NT agonists would require lower doses to produce significant analgesic effects, hence decreasing opioid-induced adverse effects. We used isobologram analyses to determine if the combination of a NT brain-penetrant analog, An2-NT(8-13) with morphine results in an inhibitory, synergistic or additive analgesic response. We found that intravenous administration of An2-NT(8-13) reduced by 90% the nocifensive behaviors induced by formalin injection, at the dose of 0.018 mg/kg. Likewise, subcutaneous morphine reduced pain by 90% at 1.8 mg/kg. Importantly, isobologram analyses revealed that the co-injection of An2-NT(8-13) with morphine induced an additive analgesic response. We finally assessed the effects of morphine and An2-NT(8-13) on the gastrointestinal tract motility using the charcoal meal test. As opposed to morphine which significantly reduced the intestinal motility at the analgesic effective dose of 1.8 mg/kg, An2-NT(8-13) did not affect the charcoal meal intestinal transit at 0.018 mg/kg. Interestingly, at the dose providing 90% pain relief, the co-administration of morphine with An2-NT(8-13) had a reduced effect on constipation. Altogether, these results suggest that combining NT agonists with morphine may improve its analgesic/adverse effect ratio.


Assuntos
Analgésicos Opioides/administração & dosagem , Neurotensina/administração & dosagem , Medição da Dor/efeitos dos fármacos , Fragmentos de Peptídeos/administração & dosagem , Peptídeos/administração & dosagem , Receptores de Neurotensina/agonistas , Receptores Opioides mu/agonistas , Animais , Relação Dose-Resposta a Droga , Quimioterapia Combinada , Masculino , Morfina/administração & dosagem , Medição da Dor/métodos , Ratos , Ratos Sprague-Dawley , Receptores de Neurotensina/metabolismo , Receptores Opioides mu/metabolismo
8.
J Cell Physiol ; 234(3): 2851-2865, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30132865

RESUMO

High nuclear expression of G protein-coupled receptors, including kinin B1 receptors (B1R), has been observed in several human cancers, but the clinical significance of this is unknown. We put forward the hypothesis that these "nuclearized" kinin B1R contribute to tumorigenicity and can be a new target in anticancer strategies. Our initial immunostaining and ultrastructural electron microscopy analyses demonstrated high B1R expression predominantly located at internal/nuclear compartments in the MDA-MB-231 triple-negative breast cancer (TNBC) cell line as well as in clinical samples of patients with TNBC. On the basis of these findings, in the present study, we evaluated the anticancer therapeutic potential of newly identified, cell-permeable B1R antagonists in MDA-MB-231 cells (ligand-receptor binding/activity assays and LC-MS/MS analyses). We found that these compounds (SSR240612, NG67, and N2000) were more toxic to MDA-MB-231 cells in comparison with low- or non-B1R expressing MCF-10A normal human mammary epithelial cells and COS-1 cells, respectively (clonogenic, MTT proliferative/cytocidal assays, and fluorescence-activated cell-sorting (FACS)-based apoptosis analyses). By comparison, the peptide B1R antagonist R954 unable to cross cell membrane failed to produce anticancer effects. Furthermore, the putative mechanisms underlying the anticancer activities of cell-penetrant B1R antagonists were assessed by analyzing cell cycle regulation and signaling molecules related to cell survival and apoptosis (FACS and western blot). Finally, drug combination experiments showed that cell-penetrant B1R antagonists can cooperate with suboptimal doses of chemotherapeutic agents (doxorubicin and paclitaxel) to promote TNBC death. This study provides evidence on the potential value of internally acting kinin B1R antagonists in averting growth of breast cancer.


Assuntos
Peptídeos Penetradores de Células/farmacologia , Receptor B1 da Bradicinina/genética , Receptores Acoplados a Proteínas G/genética , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Antagonistas de Receptor B1 da Bradicinina/farmacologia , Células COS , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular/efeitos dos fármacos , Permeabilidade da Membrana Celular/genética , Chlorocebus aethiops , Doxorrubicina/farmacologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Paclitaxel/farmacologia , Transdução de Sinais/efeitos dos fármacos , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia
9.
Pharmacol Res ; 131: 7-16, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29530600

RESUMO

The apelinergic system is an important player in the regulation of both vascular tone and cardiovascular function, making this physiological system an attractive target for drug development for hypertension, heart failure and ischemic heart disease. Indeed, apelin exerts a positive inotropic effect in humans whilst reducing peripheral vascular resistance. In this study, we investigated the signaling pathways through which apelin exerts its hypotensive action. We synthesized a series of apelin-13 analogs whereby the C-terminal Phe13 residue was replaced by natural or unnatural amino acids. In HEK293 cells expressing APJ, we evaluated the relative efficacy of these compounds to activate Gαi1 and GαoA G-proteins, recruit ß-arrestins 1 and 2 (ßarrs), and inhibit cAMP production. Calculating the transduction ratio for each pathway allowed us to identify several analogs with distinct signaling profiles. Furthermore, we found that these analogs delivered i.v. to Sprague-Dawley rats exerted a wide range of hypotensive responses. Indeed, two compounds lost their ability to lower blood pressure, while other analogs significantly reduced blood pressure as apelin-13. Interestingly, analogs that did not lower blood pressure were less effective at recruiting ßarrs. Finally, using Spearman correlations, we established that the hypotensive response was significantly correlated with ßarr recruitment but not with G protein-dependent signaling. In conclusion, our results demonstrated that the ßarr recruitment potency is involved in the hypotensive efficacy of activated APJ.


Assuntos
Anti-Hipertensivos/farmacologia , Receptores de Apelina/metabolismo , Pressão Sanguínea/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , beta-Arrestinas/metabolismo , Animais , Anti-Hipertensivos/química , AMP Cíclico/metabolismo , Células HEK293 , Humanos , Hipotensão/tratamento farmacológico , Hipotensão/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/química , Masculino , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos
10.
Oncotarget ; 9(11): 9885-9906, 2018 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-29515778

RESUMO

G protein-coupled receptors (GPCRs) are integral cell-surface proteins having a central role in tumor growth and metastasis. However, several GPCRs retain an atypical intracellular/nuclear location in various types of cancer. The pathological significance of this is currently unknown. Here we extend this observation by showing that the bradykinin B2R (BK-B2R) is nuclearly expressed in the human triple-negative breast cancer (TNBC) cell line MDA-MB-231 and in human clinical specimens of TNBC. We posited that these "nuclearized" receptors could be involved in oncogenic signaling linked to aberrant growth and survival maintenance of TNBC. We used cell-penetrating BK-B2R antagonists, including FR173657 and novel transducible, cell-permeable forms of the peptide B2R antagonist HOE 140 (NG68, NG134) to demonstrate their superior efficacy over impermeable ones (HOE 140), in blocking proliferation and promoting apoptosis of MDA-MB-231 cells. Some showed an even greater antineoplastic activity over conventional chemotherapeutic drugs in vitro. The cell-permeable B2R antagonists had less to no anticancer effects on B2R shRNA-knockdown or non-B2R expressing (COS-1) cells, indicating specificity in their action. Possible mechanisms of their anticancer effects may involve activation of p38kinase/p27Kip1 pathways. Together, our data support the existence of a possible intracrine signaling pathway via internal/nuclear B2R, critical for the growth of TNBC cells, and identify new chemical entities that enable to target the corresponding intracellular GPCRs.

11.
Crit Care Med ; 45(11): e1139-e1148, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28777197

RESUMO

OBJECTIVES: Apelin-13 was recently proposed as an alternative to the recommended ß-adrenergic drugs for supporting endotoxin-induced myocardial dysfunction. Since Apelin-13 signals through its receptor (Apelin peptide jejunum) to exert singular inotropic/vasotropic actions and to optimize body fluid balance, this candidate pathway might benefit septic shock management. Whether the newly discovered ELABELA (ELA), a second endogenous ligand of the Apelin peptide jejunum receptor highly expressed in the kidney, further improves cardio-renal impairment remains unknown. DESIGN, SETTING, AND SUBJECTS: Interventional study in a rat model of septic shock (128 adult males) to assess the effects of ELA and Apelin-13 on vascular and cardio-renal function. Experiments were performed in a tertiary care University-based research institute. INTERVENTIONS: Polymicrobial sepsis-induced cardiac dysfunction was produced by cecal ligation puncture to assess hemodynamic efficacy, cardioprotection, and biomechanics under acute or continuous infusions of the apelinergic agonists ELA or Apelin-13 (39 and 15 µg/kg/hr, respectively) versus normal saline. MEASUREMENTS AND MAIN RESULTS: Apelinergic agonists improved 72-hour survival after sepsis induction, with ELA providing the best clinical outcome after 24 hours. Apelinergic agonist infusion counteracted cecal ligation puncture-induced myocardial dysfunction by improving left ventricular pressure-volume relationship. ELA-treated cecal ligation puncture rats were the only group to 1) display a significant improvement in left ventricular filling as shown by increased E-wave velocity and left ventricular end-diastolic volume, 2) exhibit a higher plasma volume, and 3) limit kidney injury and free-water clearance. These beneficial renal effects were superior to Apelin-13, likely because full-length ELA enabled a distinctive regulation of pituitary vasopressin release. CONCLUSIONS: Activation of the apelinergic system by exogenous ELA or Apelin-13 infusion improves cardiovascular function and survival after cecal ligation puncture-induced sepsis. However, ELA proved better than Apelin-13 by improving fluid homeostasis, cardiovascular hemodynamics recovery, and limiting kidney dysfunction in a vasopressinergic-dependent manner.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Hormônios Peptídicos/farmacologia , Choque Séptico/tratamento farmacológico , Animais , Biomarcadores , Citocinas/imunologia , Modelos Animais de Doenças , Ecocardiografia , Hemodinâmica/efeitos dos fármacos , Masculino , Ratos , Reação em Cadeia da Polimerase em Tempo Real
12.
J Med Chem ; 59(7): 2962-72, 2016 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-26986036

RESUMO

ELABELA (ELA) was recently discovered as a novel endogenous ligand of the apelin receptor (APJ), a G protein-coupled receptor. ELA signaling was demonstrated to be crucial for normal heart and vasculature development during embryogenesis. We delineate here ELA's structure-activity relationships and report the identification of analogue 3 (ELA(19-32)), a fragment of ELA that binds to APJ, activates the Gαi1 and ß-arrestin-2 signaling pathways, and induces receptor internalization similarly to its parent endogenous peptide. An alanine scan performed on 3 revealed that the C-terminal residues are critical for binding to APJ and signaling. Finally, using isolated-perfused hearts and in vivo hemodynamic and echocardiographic measurements, we demonstrate that ELA and 3 both reduce arterial pressure and exert positive inotropic effects on the heart. Altogether, these results present ELA and 3 as potential therapeutic options in managing cardiovascular diseases.


Assuntos
Fármacos Cardiovasculares/farmacologia , Fragmentos de Peptídeos/farmacologia , Hormônios Peptídicos/química , Relação Estrutura-Atividade , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Receptores de Apelina , Pressão Sanguínea/efeitos dos fármacos , Fármacos Cardiovasculares/química , Eletrocardiografia , Coração/efeitos dos fármacos , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/fisiopatologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Masculino , Dados de Sequência Molecular , Técnicas de Cultura de Órgãos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Hormônios Peptídicos/metabolismo , Hormônios Peptídicos/farmacologia , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo
13.
Biol Chem ; 397(4): 365-72, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26565554

RESUMO

Several studies have shown the potential therapeutic utility of kinin B1 receptor (B1R) peptide agonists in neurological and ischemic cardiovascular diseases and brain cancer. Preclinical safety studies are a prerequisite for further drug development. The objectives of this study were to determine the acute toxicity and pharmacokinetics of the peptide B1R agonist, SarLys[dPhe8]desArg9-bradykinin (NG29), as trifluoroacetate (TFacetate) or acetate salt form, following intravenous injection in rats. A maximum tolerated dose (MTD) of NG29-TFacetate was established at 75 mg/kg from the results of a dose range-finding study (up to 200 mg/kg). The short-term (4-day) repeat-dose toxicity study of NG29, using its MTD value, showed that NG29-acetate exhibited minimal non-adverse clinical pathology changes in hematology, coagulation, clinical chemistry and urine parameters and severe kidney histopathological changes characterized by renal tubular degeneration. No such effects were observed with NG29-TFacetate. At the injection site, NG29-TFacetate was considered to be more locally irritating when compared to the acetate form. The extent of exposure and half-life values of NG29-TFacetate were comparable to the acetate form (AUC0-α of 10.2 mg/l*h vs. 9.9 mg/l*h; T1/2 of 2.3 h vs. 2.4 h). This study shows that in rats NG29-TFacetate exhibits a superior tolerability profile compared with the peptide acetate form.


Assuntos
Peptídeos/efeitos adversos , Peptídeos/farmacocinética , Receptor B1 da Bradicinina/agonistas , Animais , Relação Dose-Resposta a Droga , Células HEK293 , Meia-Vida , Humanos , Injeções Intravenosas , Masculino , Dose Máxima Tolerável , Peptídeos/administração & dosagem , Peptídeos/química , Ratos , Ratos Wistar
14.
J Med Chem ; 58(5): 2431-40, 2015 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-25668242

RESUMO

Apelin is the endogenous ligand of the APJ receptor, a member of the G protein-coupled receptor family. This system plays an important role in the regulation of blood pressure and cardiovascular functions. To better understand the role of its C-terminal Phe(13) residue on ligand binding, receptor signaling, and hypotension, we report a series of modified analogues in which Phe(13) was substituted by unnatural amino acids. These modifications delivered new compounds exhibiting higher affinity and potency to inhibit cAMP accumulation compared to apelin-13. In particular, analogues Bpa(13) or (α-Me)Phe(13) were 30-fold more potent to inhibit cAMP accumulation than apelin-13. Tyr(OBn)(13) substitution led to a 60-fold improvement in binding affinity and induced stronger and more sustained drop in blood pressure compared to apelin-13. Our study identified new potent analogues of apelin-13, which represent valuable probes to better understand its structure-function relationship.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , AMP Cíclico/metabolismo , Hipotensão/tratamento farmacológico , Peptídeos e Proteínas de Sinalização Intercelular/química , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Substituição de Aminoácidos , Animais , Apelina , Ligantes , Masculino , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Relação Estrutura-Atividade
15.
Cancer Biol Ther ; 14(9): 806-11, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23792591

RESUMO

The low permeability of the BBB is largely responsible for the lack of effective systemic chemotherapy against primary and metastatic brain tumors. Kinin B1R and B2R have been shown to mediate reversible tumor-selective BBB disruption in preclinical animal models. We investigated whether co-administration of two novel potent kinin B1R and B2R agonists offers an advantage over administering each agonist alone for enhancing BBB permeability and tumor targeting of drugs in the malignant F98 glioma rat model. A new covalent kinin heterodimer that equally stimulates B1R and B2R was also constructed for the purpose of our study. We found that co-administration of B1R and B2R agonists, or alternatively administration of the kinin heterodimer more effectively delivered the MRI contrast agent Gd-DTPA and the anticancer drug carboplatin to brain tumors and surrounding tissues than the agonists alone (determined by MRI and ICP-MS methods). Importantly, the efficient delivery of carboplatin by the dual kinin receptor targeting on the BBB translated into increased survival of glioma-bearing rats. Thus, this report describes a potential strategy for maximizing the brain bioavailability and therapeutic efficacy of chemotherapeutic drugs.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Neoplasias Encefálicas/tratamento farmacológico , Glioma/tratamento farmacológico , Cininas/farmacologia , Receptor B1 da Bradicinina/agonistas , Receptor B2 da Bradicinina/agonistas , Animais , Antineoplásicos/administração & dosagem , Barreira Hematoencefálica/metabolismo , Neoplasias Encefálicas/metabolismo , Carboplatina/administração & dosagem , Química Click , Meios de Contraste/administração & dosagem , Dimerização , Sinergismo Farmacológico , Glioma/metabolismo , Células HEK293 , Humanos , Cininas/química , Permeabilidade , Ratos Endogâmicos F344 , Receptor B1 da Bradicinina/metabolismo , Receptor B2 da Bradicinina/metabolismo
16.
PLoS One ; 7(5): e37485, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22629405

RESUMO

Treatment of malignant glioma with chemotherapy is limited mostly because of delivery impediment related to the blood-brain tumor barrier (BTB). B1 receptors (B1R), inducible prototypical G-protein coupled receptors (GPCR) can regulate permeability of vessels including possibly that of brain tumors. Here, we determine the extent of BTB permeability induced by the natural and synthetic peptide B1R agonists, LysdesArg(9)BK (LDBK) and SarLys[dPhe(8)]desArg(9)BK (NG29), in syngeneic F98 glioma-implanted Fischer rats. Ten days after tumor inoculation, we detected the presence of B1R on tumor cells and associated vasculature. NG29 infusion increased brain distribution volume and uptake profiles of paramagnetic probes (Magnevist and Gadomer) at tumoral sites (T(1)-weighted imaging). These effects were blocked by B1R antagonist and non-selective cyclooxygenase inhibitors, but not by B2R antagonist and non-selective nitric oxide synthase inhibitors. Consistent with MRI data, systemic co-administration of NG29 improved brain tumor delivery of Carboplatin chemotherapy (ICP-Mass spectrometry). We also detected elevated B1R expression in clinical samples of high-grade glioma. Our results documented a novel GPCR-signaling mechanism for promoting transient BTB disruption, involving activation of B1R and ensuing production of COX metabolites. They also underlined the potential value of synthetic biostable B1R agonists as selective BTB modulators for local delivery of different sized-therapeutics at (peri)tumoral sites.


Assuntos
Bradicinina/análogos & derivados , Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Calidina/análogos & derivados , Receptor B1 da Bradicinina/agonistas , Adulto , Idoso , Animais , Transporte Biológico/efeitos dos fármacos , Bradicinina/farmacologia , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/tratamento farmacológico , Permeabilidade Capilar/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Glioma/irrigação sanguínea , Glioma/tratamento farmacológico , Humanos , Calidina/farmacologia , Masculino , Pessoa de Meia-Idade , Permeabilidade/efeitos dos fármacos , Ratos , Ratos Endogâmicos F344 , Receptor B1 da Bradicinina/metabolismo
17.
Neuropeptides ; 44(2): 177-85, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20080302

RESUMO

Treatment of malignant glioma with chemotherapy is limited mostly because of delivery impediment related to the blood-brain barrier (BBB). One approach for transporting drugs across the BBB involves the activation of bradykinin-B2 receptors (BK-B2R). Our objective was to pharmacologically characterize the BBB permeability induced by the synthetic biostable BK-B2R analogue [Phe(8)psi(CH(2)NH)Arg(9)]-BK (R523) in F98 glioma-implanted Fischer rats. On day 10 post-inoculation, we detected the presence of B2R in the tumor cells and the peritumoral microvasculature (RT-PCR and immunohistochemistry). We assessed BBB permeability before and after the intracarotid (i.c.) infusion of R523 (0.1ml/min for 5min; 2.5, 10, and 50nmol/kg/min) using non-invasive dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) with the different sized-contrast agents Gd-DTPA (0.5kDa) and Gadomer (17kDa) (0.25mmol/kg via the caudal vein). T(1)-weighted images were analyzed for the presence or absence of contrast enhancement within and surrounding the tumor area and mathematically processed to yield a contrast agent distribution volume (CADV), which was used as an indicator of vascular permeability. Our results showed that the agonist R523 increased, in a dose-dependent manner, the CADV indexes of Gd-DTPA and Gadomer, with a maximum 2-fold increase in brain uptake of both CA. The increase in CADV induced by R523 (10nmol/kg/min) was prevented by the B2R antagonist HOE140 (20nmol/kg/min, i.c.) and the nitric oxide synthase inhibitor L-NA (5mg/kg, i.v.) but not by the B1R antagonist R892 (20nmol/kg/min, i.c.) or the cyclooxygenase inhibitor Meclofenamate (5mg/kg, i.v.). The BBB permeabilizing effect of R523 (10nmol/kg/min) lasted for <1h and was accompanied by a dose-related fall in arterial blood pressure. We concluded that R523 allows the extravasation of hydrophilic macromolecular agents (17kDa) into tumor tissues by inducing selective tumor BBB permeability via B2R- and NO-dependent mechanisms.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Bradicinina/análogos & derivados , Neoplasias Encefálicas/metabolismo , Permeabilidade Capilar/fisiologia , Glioma/metabolismo , Receptor B2 da Bradicinina/metabolismo , Animais , Pressão Sanguínea/fisiologia , Barreira Hematoencefálica/metabolismo , Bradicinina/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Glioma/tratamento farmacológico , Imuno-Histoquímica , Imageamento por Ressonância Magnética , Masculino , Transplante de Neoplasias , Óxido Nítrico/metabolismo , Ratos , Ratos Endogâmicos F344 , Receptor B2 da Bradicinina/agonistas , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
Peptides ; 30(4): 788-95, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19150636

RESUMO

There is some evidence to suggest that inducible kinin B1 receptors (B1R) may play beneficial and protecting roles in cardiovascular-related pathologies such as hypertension, diabetes, and ischemic organ diseases. Peptide B1R agonists bearing optimized pharmacological features (high potency, selectivity and stability toward proteolysis) hold promise as valuable therapeutic agents in the treatment of these diseases. In the present study, we used solid-phase methodology to synthesize a series of novel peptide analogues based on the sequence of Sar[dPhe(8)]desArg(9)-bradykinin, a relatively stable peptide agonist with moderate affinity for the human B1R. We evaluated the pharmacological properties of these peptides using (1) in vitro competitive binding experiments on recombinant human B1R and B2R (for index of selectivity determination) in transiently transfected human embryonic kidney 293 cells (HEK-293T cells), (2) ex vivo vasomotor assays on isolated human umbilical veins expressing endogenous human B1R, and (3) in vivo blood pressure tests using anesthetized lipopolysaccharide-immunostimulated rabbits. Key chemical modifications at the N-terminus, the positions 3 and 5 on Sar[dPhe(8)]desArg(9)-bradykinin led to potent analogues. For example, peptides 18 (SarLys[Hyp(3),Cha(5), dPhe(8)]desArg(9)-bradykinin) and 20 (SarLys[Hyp(3),Igl(5), dPhe(8)]desArg(9)-bradykinin) outperformed the parental molecule in terms of affinity, functional potency and duration of action in vitro and in vivo. These selective agonists should be valuable in future animal and human studies to investigate the potential benefits of B1R activation.


Assuntos
Peptídeos/farmacologia , Receptores da Bradicinina/agonistas , Animais , Ligação Competitiva , Pressão Sanguínea/efeitos dos fármacos , Linhagem Celular , Humanos , Técnicas In Vitro , Lipopolissacarídeos/administração & dosagem , Masculino , Peptídeos/química , Peptídeos/metabolismo , Conformação Proteica , Coelhos , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley
19.
Peptides ; 30(4): 777-87, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19111586

RESUMO

The nonapeptide bradykinin (BK) is involved in the genesis of inflammation, edema and in pain mediation. As such, much effort has gone into the development of peptide/non-peptide antagonists to counteract these processes. However, there is an increasing awareness of the potential value of chemically stable BK agonists in the treatment of diabetes and cardiovascular diseases. In this study, a structure-activity relationship study of BK was performed to develop potent and stable peptide mimetics active at the human B2 receptors (hB2R). Twenty-three analogues were produced with substitutions at positions 1, 3, 5, 7, 8 and/or 9 of BK. In vitro binding (on transiently transfected HEK-293T cells) and biological activities (vasomotricity tests on human umbilical veins, MAPK assays on HEK-293T cells) of novel BK peptide derivatives at hB2R were determined alongside with previously reported synthetic agonists (e.g. RMP-7, JMV1609, FR190997). Some peptides were also tested in vivo in rats and rabbits using blood pressure assays. Two compounds, [Hyp(3), Thi(5), Cha(8)]-BK and [Hyp(3), Thi(5), (N)Chg(7), Thi(8)]-BK, exhibited equivalent (or even greater) in vitro affinities and potencies to BK at the naturally expressed and recombinant hB2R. Their potency and duration of action in vivo were highly superior to BK, thus inferring that they can withstand intravascular proteolysis. These novel compounds show promise as candidates for investigating the pharmacology of BK receptors and developing potential therapeutical applications.


Assuntos
Bradicinina/química , Peptídeos/farmacologia , Receptor B2 da Bradicinina/agonistas , Animais , Pressão Sanguínea/efeitos dos fármacos , Western Blotting , Linhagem Celular , Humanos , Masculino , Fosforilação , Coelhos , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Relação Estrutura-Atividade
20.
J Cell Physiol ; 216(1): 234-44, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18264983

RESUMO

Bradykinin (BK) represents a pro-inflammatory mediator that partakes in many inflammatory diseases. The mechanism of action of BK is thought to be primarily mediated by specific cell surface membrane B2 receptors (B2Rs). Some evidence has suggested, however, the existence of an intracellular/nuclear B2R population. Whether these receptors are functional and contribute to BK signaling remains to be determined. In this study, by mean of Western blotting, 3D-confocal microscopy, receptor autoradiography and radioligand binding analysis, we showed that plasma membrane and highly purified nuclei from isolated rat hepatocytes contain specific B2R that bind BK. The results depicting B2R nuclear expression in isolated nuclear organelles were reproduced in situ on hepatic sections by immunogold labeling and transmission electron microscopy. Functional tests on single nuclei, by means of confocal microscopy and the calcium-sensitive probe fluo-4AM, showed that BK induces concentration-dependent transitory mobilization of nucleoplasmic calcium; these responses were blocked by B2R antagonist HOE 140, not by the B1R antagonist R954 and, were also found in wild-type C57/Bl6 mice, but not in B2R-KO mice. In isolated nuclei, BK elicited activation/phosphorylation of Akt, acetylation of histone H3 and ensuing pro-inflammatory iNOS gene induction as determined by Western blot and RT-PCR. ChIP assay confirmed binding of acetylated-histone H3 complexes, but not B2R, to promoter region of iNOS gene suggesting that B2R-mediated gene expression is bridged with accessory downstream effectors. This study discloses a previously undescribed mechanism in BK-induced transcriptional events, via intracrine B2R-mediated signaling, occurring in rat autologous hepatic cells.


Assuntos
Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Regulação da Expressão Gênica , Genes Precoces , Receptor B2 da Bradicinina/metabolismo , Transdução de Sinais/fisiologia , Animais , Bradicinina/metabolismo , Hepatócitos/citologia , Hepatócitos/metabolismo , Humanos , Cininogênio de Alto Peso Molecular/genética , Cininogênio de Alto Peso Molecular/metabolismo , Cininogênio de Baixo Peso Molecular/genética , Cininogênio de Baixo Peso Molecular/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Receptor B2 da Bradicinina/genética , Ativação Transcricional , Vasodilatadores/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA