Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta Gen Subj ; 1867(4): 130322, 2023 04.
Article in English | MEDLINE | ID: mdl-36773726

ABSTRACT

BACKGROUND: Ceefourin-1 is a specific MRP4/ABCC4 inhibitor with potential antileukemic activity. In this study, we evaluate the ability of ceefourin-1 alone or in combination with histamine, an approved antileukemic agent, to induce cell differentiation or apoptosis in human acute myeloid leukemic cells. We also examine ceefourin-1 toxicity in mice. METHODS: U937, HL-60, and KG1a cells were used as models for human acute myeloid leukemia. Cyclic AMP efflux was estimated by measuring intracellular and extracellular cAMP levels. Cell differentiation was assessed by levels of CD14 and CD11b by FACS, and CD88 by western blot, and by cell morphology. Apoptosis was evaluated by cleavage of caspase-3 and PARP by western blot, and by annexin V binding assay. Subacute toxicity study of ceefourin-1 was carried out in BALB/c mice. RESULTS: Ceefourin-1 inhibits cAMP exclusion in AML cells and promotes intracellular signaling via CREB. Ceefourin-1 leads AML cells to apoptosis and histamine potentiates this effect, without evidence of cell differentiation. Intraperitoneal administration of ceefourin-1 shows no important alterations in mice blood parameters, hepatic, and renal functions, nor signs of histologic damage. CONCLUSIONS: These results show that ceefourin-1 promotes apoptosis in AML cells that is enhanced by histamine. GENERAL SIGNIFICANCE: This work indicates that ceefourin-1 represents a promising molecule that could be used alone or in combination with histamine for in vivo evaluation in acute myeloid leukemia malignancies.


Subject(s)
Histamine , Leukemia, Myeloid, Acute , Animals , Humans , Mice , Apoptosis , ATP-Binding Cassette Transporters , Histamine/pharmacology , Leukemia, Myeloid, Acute/metabolism , Multidrug Resistance-Associated Proteins
2.
Pharmacol Res Perspect ; 10(2): e00913, 2022 04.
Article in English | MEDLINE | ID: mdl-35184416

ABSTRACT

G protein-coupled receptors kinase 2 (GRK2) plays a major role in receptor regulation and, as a consequence, in cell biology and physiology. GRK2-mediated receptor desensitization is performed by its kinase domain, which exerts receptor phosphorylation promoting G protein uncoupling and the cessation of signaling, and by its RGS homology (RH) domain, able to interrupt G protein signaling. Since GRK2 activity is exacerbated in several pathologies, many efforts to develop inhibitors have been conducted. Most of them were directed toward GRK2 kinase activity and showed encouraging results on in vitro systems and animal models. Nevertheless, limitations including unspecific effects or pharmacokinetics issues prevented them from advancing to clinical trials. Surprisingly, even though the RH domain demonstrated the ability to desensitize GPCRs, this domain has been less explored. Herein, we show in vitro activity of a series of compounds that, by inhibiting GRK2 RH domain, increase receptor cAMP response, avoid GRK2 translocation to the plasma membrane, inhibit coimmunoprecipitation of GRK2 with Gαs subunit of heterotrimeric G protein, and prevent receptor desensitization. Also, we preliminarily evaluated candidates' ADMET properties and observed suitable lipophilicity and cytotoxicity. These novel inhibitors of phosphorylation-independent actions of GRK2 might be useful in elucidating other RH domain roles and lay the foundation for the development of innovative pharmacologic therapy for diseases where GRK2 activity is exacerbated.


Subject(s)
Cyclic AMP/metabolism , G-Protein-Coupled Receptor Kinase 2/antagonists & inhibitors , Cell Line, Tumor , Drug Development , G-Protein-Coupled Receptor Kinase 2/metabolism , HEK293 Cells , Humans , Phosphorylation , Protein Domains/drug effects , RGS Proteins/metabolism , Signal Transduction/drug effects
3.
Eur J Pharmacol ; 896: 173913, 2021 Apr 05.
Article in English | MEDLINE | ID: mdl-33508282

ABSTRACT

Histamine H1 receptor ligands used clinically as antiallergics rank among the most widely prescribed and over-the-counter drugs in the world. They exert the therapeutic actions by blocking the effects of histamine, due to null or negative efficacy towards Gαq-phospholipase C (PLC)-inositol triphosphates (IP3)-Ca2+ and nuclear factor-kappa B cascades. However, there is no information regarding their ability to modulate other receptor responses. The aim of the present study was to investigate whether histamine H1 receptor ligands could display positive efficacy concerning receptor desensitization, internalization, signaling through Gαq independent pathways or even transcriptional regulation of proinflammatory genes. While diphenhydramine, triprolidine and chlorpheniramine activate ERK1/2 (extracellular signal-regulated kinase 1/2) pathway in A549 cells, pre-treatment with chlorpheniramine or triprolidine completely desensitize histamine H1 receptor mediated Ca2+ response, and both diphenhydramine and triprolidine lead to receptor internalization. Unlike histamine, histamine H1 receptor desensitization and internalization induced by antihistamines prove to be independent of G protein-coupled receptor kinase 2 (GRK2) phosphorylation. Also, unlike the reference agonist, the recovery of the number of cell-surface histamine H1 receptors is a consequence of de novo synthesis. On the other hand, all of the ligands lack efficacy regarding cyclooxygenase-2 (COX-2) and interleukin-8 (IL-8) mRNA regulation. However, a prolonged exposure with each of the antihistamines impaires the increase in COX-2 and IL-8 mRNA levels induced by histamine, even after ligand removal. Altogether, these findings demonstrate the biased nature of histamine H1 receptor ligands contributing to a more accurate classification, and providing evidence for a more rational and safe use of them.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Histamine Agonists/pharmacology , Histamine H1 Antagonists/pharmacology , Receptors, Histamine H1/drug effects , A549 Cells , Calcium Signaling/drug effects , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Drug Inverse Agonism , Enzyme Activation , G-Protein-Coupled Receptor Kinase 2/genetics , G-Protein-Coupled Receptor Kinase 2/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Humans , Inflammation Mediators/metabolism , Inositol 1,4,5-Trisphosphate/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Ligands , Phosphorylation , Protein Transport , Receptors, Histamine H1/metabolism , Type C Phospholipases/metabolism
4.
FEBS J ; 288(1): 229-243, 2021 01.
Article in English | MEDLINE | ID: mdl-32333821

ABSTRACT

Intracellular cAMP (i-cAMP) levels play an important role in acute myeloid leukemia (AML) cell proliferation and differentiation. Its levels are the result of cAMP production, degradation, and exclusion. We have previously described histamine H2 receptors and MRP4/ABCC4 as two potential targets for AML therapy. Acting through histamine H2 receptors, histamine increases cAMP production/synthesis, while MRP4/ABCC4 is responsible for the exclusion of this cyclic nucleotide. In this study, we show that histamine treatment induces MRP4/ABCC4 expression, augmenting cAMP efflux, and that histamine, in combination with MRP inhibitors, is able to reduce AML cell proliferation. Histamine, through histamine H2 receptor, increases i-cAMP levels and induces MRP4 transcript and protein levels in U937, KG1a, and HL-60 cells. Moreover, histamine induces MRP4 promoter activity in HEK293T cells transfected with histamine H2 receptor (HEK293T-H2 R). Our results support that the cAMP/Epac-PKA pathway, and not MEK/ERK nor PI3K/AKT signaling cascades, is involved in histamine-mediated upregulation of MRP4 levels. Finally, the addition of histamine potentiates the inhibition of U937, KG1a, and HL-60 cell proliferation induced by MRP4 inhibitors. Our data highlight that the use of a poly-pharmacological approach aimed at different molecular targets would be beneficial in AML treatment.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP/metabolism , Guanine Nucleotide Exchange Factors/genetics , Histamine/pharmacology , Multidrug Resistance-Associated Proteins/genetics , Receptors, Histamine H2/genetics , Benzothiazoles/pharmacology , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation, Leukemic , Genes, Reporter , Guanine Nucleotide Exchange Factors/metabolism , HEK293 Cells , HL-60 Cells , Histamine/metabolism , Humans , Luciferases/genetics , Luciferases/metabolism , Molecular Targeted Therapy/methods , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Multidrug Resistance-Associated Proteins/metabolism , Probenecid/pharmacology , Promoter Regions, Genetic , Propionates/pharmacology , Quinolines/pharmacology , Receptors, Histamine H2/metabolism , Signal Transduction , Triazoles/pharmacology , U937 Cells
5.
Antiviral Res ; 179: 104817, 2020 07.
Article in English | MEDLINE | ID: mdl-32387475

ABSTRACT

Respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract disease and bronchiolitis in children, as well as an important cause of morbidity and mortality in elderly and immunocompromised individuals. However, there is no safe and efficacious RSV vaccine or antiviral treatment. Toll Like Receptors (TLR) are important molecular mediators linking innate and adaptive immunity, and their stimulation by cognate agonists has been explored as antiviral agents. Imiquimod is known as a TLR7 agonist, but additionally acts as an antagonist for adenosine receptors. In this study, we demonstrate that imiquimod, but not resiquimod, has direct anti-RSV activity via PKA pathway in HEp-2 and A549 cells, independently of an innate response. Imiquimod restricts RSV infection after viral entry into the host cell, interfering with viral RNA and protein synthesis. Probably as a consequence of these anti-RSV properties, imiquimod displays cytokine modulating activity in RSV infected epithelial cells. Moreover, in a murine model of RSV infection, imiquimod treatment improves the course of acute disease, evidenced by decreased weight loss, reduced RSV lung titers, and attenuated airway inflammation. Consequently, imiquimod represents a promising therapeutic alternative against RSV infection and may inform the development of novel therapeutic targets to control RSV pathogenesis.


Subject(s)
Antiviral Agents/therapeutic use , Imiquimod/therapeutic use , Inflammation/drug therapy , Respiratory Syncytial Virus Infections/immunology , Signal Transduction , Virus Replication/drug effects , A549 Cells , Animals , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytokines/antagonists & inhibitors , Cytokines/immunology , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/virology , Female , Humans , Inflammation/virology , Lung/drug effects , Lung/virology , Mice , Mice, Inbred BALB C , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Virus, Human/physiology , Viral Load
6.
Front Pharmacol ; 11: 113, 2020.
Article in English | MEDLINE | ID: mdl-32153413

ABSTRACT

G protein coupled receptor (GPCR) kinases (GRKs) are key regulators of GPCR signaling. Canonical mechanism of GPCR desensitization involves receptor phosphorylation by GRKs followed by arrestin recruitment and uncoupling from heterotrimeric G protein. Although ß3-adrenergic receptor (ß3AR) lacks phosphorylation sites by GRKs, agonist treatment proved to induce ß3AR desensitization in many cell types. Here we show that GRK2 mediates short-term desensitization of ß3AR by a phosphorylation independent mechanism but mediated by its domain homologous to the regulator of G protein signaling (RGS). HEK293T cells overexpressing human ß3AR presented a short-term desensitization of cAMP response stimulated by the ß3AR agonist, BRL37344, and not by forskolin. We found that ß3AR desensitization was higher in cells co-transfected with GRK2. Similarly, overexpression of the RGS homology domain but not kinase domain of GRK2 increased ß3AR desensitization. Consistently, stimulation of ß3AR increased interaction between GRK2 and Gαs subunit. Furthermore, in rat cardiomyocytes endogenously expressing ß3AR, transfection with dominant negative mutant of RH domain of GRK2 (GRK2/D110A) increased cAMP response to BRL37344 and inhibited receptor desensitization. We expect our study to be a starting point for more sophisticated characterization of the consequences of GRK2 mediated desensitization of the ß3AR in heart function and disease.

7.
Pharmacol Res Perspect ; 7(6): e00531, 2019 12.
Article in English | MEDLINE | ID: mdl-31687162

ABSTRACT

Glucocorticoids are among the most effective drugs to treat asthma. However, the severe adverse effects associated generate the need for its therapeutic optimization. Conversely, though histamine is undoubtedly related to asthma development, there is a lack of efficacy of antihistamines in controlling its symptoms, which prevents their clinical application. We have reported that antihistamines potentiate glucocorticoids' responses in vitro and recent observations have indicated that the coadministration of an antihistamine and a synthetic glucocorticoid has synergistic effects on a murine model of allergic rhinitis. Here, the aim of this work is to establish if this therapeutic combination could be beneficial in a murine model of asthma. We used an allergen-induced model of asthma (employing ovalbumin) to evaluate the effects of the synthetic glucocorticoid dexamethasone combined with the antihistamine azelastine. Our results indicate that the cotreatment with azelastine and a suboptimal dose of dexamethasone can improve allergic lung inflammation as shown by a decrease in eosinophils in bronchoalveolar lavage, fewer peribronchial and perivascular infiltrates, and mucin-producing cells. In addition, serum levels of allergen-specific IgE and IgG1 were also reduced, as well as the expression of lung inflammatory-related genes IL-4, IL-5, Muc5AC, and Arginase I. The potentiation of dexamethasone effects by azelastine could allow to reduce the effective glucocorticoid dose needed to achieve a therapeutic effect. These findings provide first new insights into the potential benefits of glucocorticoids and antihistamines combination for the treatment of asthma and grants further research to evaluate this approach in other related inflammatory conditions.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Asthma/drug therapy , Dexamethasone/pharmacology , Phthalazines/pharmacology , Administration, Intranasal , Animals , Anti-Asthmatic Agents/therapeutic use , Asthma/blood , Asthma/immunology , Asthma/pathology , Dexamethasone/therapeutic use , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Synergism , Drug Therapy, Combination/methods , Female , Glucocorticoids/pharmacology , Glucocorticoids/therapeutic use , HEK293 Cells , Histamine H1 Antagonists, Non-Sedating/pharmacology , Histamine H1 Antagonists, Non-Sedating/therapeutic use , Humans , Lung/drug effects , Lung/immunology , Lung/pathology , Mice , Ovalbumin/immunology , Phthalazines/therapeutic use , Receptors, Glucocorticoid/agonists , Receptors, Glucocorticoid/metabolism , Transcriptional Activation/drug effects , Transcriptional Activation/immunology
8.
Mol Pharmacol ; 96(1): 13-25, 2019 07.
Article in English | MEDLINE | ID: mdl-31043460

ABSTRACT

Pancreatic cancer is one of the most lethal types of tumors with no effective therapy available; is currently the third leading cause of cancer in developed countries; and is predicted to become the second deadliest cancer in the United States by 2030. Due to the marginal benefits of current standard chemotherapy, the identification of new therapeutic targets is greatly required. Considering that cAMP pathway is commonly activated in pancreatic ductal adenocarcinoma (PDAC) and its premalignant lesions, we aim to investigate the multidrug resistance-associated protein 4 (MRP4)-dependent cAMP extrusion process as a cause of increased cell proliferation in human PDAC cell lines. Our results from in silico analysis indicate that MRP4 expression may influence PDAC patient outcome; thus, high MRP4 levels could be indicators of poor survival. In addition, we performed in vitro experiments and identified an association between higher MRP4 expression levels and more undifferentiated and malignant models of PDAC and cAMP extrusion capacity. We studied the antiproliferative effect and the overall cAMP response of three MRP4 inhibitors, probenecid, MK571, and ceefourin-1 in PDAC in vitro models. Moreover, MRP4-specific silencing in PANC-1 cells reduced cell proliferation (P < 0.05), whereas MRP4 overexpression in BxPC-3 cells significantly incremented their growth rate in culture (P < 0.05). MRP4 pharmacological inhibition or silencing abrogated cell proliferation through the activation of the cAMP/Epac/Rap1 signaling pathway. Also, extracellular cAMP reverted the antiproliferative effect of MRP4 blockade. Our data highlight the MRP4-dependent cAMP extrusion process as a key participant in cell proliferation, indicating that MRP4 could be an exploitable therapeutic target for PDAC. SIGNIFICANCE STATEMENT: ABCC4/MRP4 is the main transporter responsible for cAMP efflux. In this work, we show that MRP4 expression may influence PDAC patient outcome and identify an association between higher MRP4 expression levels and more undifferentiated and malignant in vitro models of PDAC. Findings prove the involvement of MRP4 in PDAC cell proliferation through a novel extracellular cAMP mitogenic pathway and further support MRP4 inhibition as a promising therapeutic strategy for PDAC treatment.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Cyclic AMP/metabolism , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Pancreatic Neoplasms/metabolism , Benzothiazoles/pharmacology , Carcinoma, Pancreatic Ductal/genetics , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Computer Simulation , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing , HEK293 Cells , Humans , Pancreatic Neoplasms/genetics , Probenecid/pharmacology , Prognosis , Propionates/pharmacology , Quinolines/pharmacology , Signal Transduction/drug effects , Survival Analysis , Triazoles/pharmacology , Up-Regulation
9.
Eur J Pharmacol ; 847: 42-52, 2019 Mar 15.
Article in English | MEDLINE | ID: mdl-30685431

ABSTRACT

Histamine [2-(4-Imidazolyl)-ethylamine] modulates different biological processes, through histamine H1 and H2 receptors, and their respective blockers are widely used in treating allergic and gastric acid-related disorders. Histamine H1 and H2 receptor crossdesensitization and cointernalization induced by its agonists have been previously described. In this study, we show how this crosstalk determines the response to histamine H1 and H2 receptor inverse agonists and how histamine H1 and H2 receptor inverse agonists interfere with the other receptor's response to agonists. By desensitization assays we demonstrate that histamine H1 and H2 receptor inverse agonists induce a crossregulation between both receptors. In this sense, the histamine H1 receptor inverse agonists desensitize the cAMP response to amthamine, a histamine H2 receptor agonist. In turn, histamine H2 receptor inverse agonists interfere with histamine H1 receptor signaling. We also determine that the crossdesensitization induced by histamine H1 or H2 receptor agonists alters the histamine inverse agonists receptor response: activation of histamine H1 receptor affects cAMP response induced by histamine H2 receptor inverse agonists, whereas histamine H2 receptor agonist induces a negative regulation on the anti-inflammatory response of histamine H1 receptor inverse agonists. Binding studies revealed that histamine H1 and H2 receptors cointernalize after stimulus with histamine receptor inverse agonists. In addition, the inhibition of the internalization process prevents receptor crossregulation. Our study provides new insights in the mechanisms of action of histamine H1 and H2 receptors that explain the effect of histamine H1 and H2 receptor inverse agonists and opens up new venues for novel therapeutic applications.


Subject(s)
Histamine Agonists/metabolism , Histamine H1 Antagonists/metabolism , Receptors, Histamine H1/metabolism , Receptors, Histamine H2/metabolism , Cell Line , Cell Line, Tumor , HEK293 Cells , Histamine/metabolism , Humans , Signal Transduction/physiology , U937 Cells
10.
Curr Med Chem ; 26(7): 1270-1307, 2019.
Article in English | MEDLINE | ID: mdl-29284392

ABSTRACT

MRP4 transports multiple endogenous and exogenous substances and is critical not only for detoxification but also in the homeostasis of several signaling molecules. Its dysregulation has been reported in numerous pathological disorders, thus MRP4 appears as an attractive therapeutic target. However, the efficacy of MRP4 inhibitors is still controversial. The design of specific pharmacological agents with the ability to selectively modulate the activity of this transporter or modify its affinity to certain substrates represents a challenge in current medicine and chemical biology. The first step in the long process of drug rational design is to identify the therapeutic target and characterize the mechanism by which it affects the given pathology. In order to develop a pharmacological agent with high specific activity, the second step is to systematically study the structure of the target and identify all the possible binding sites. Using available homology models and mutagenesis assays, in this review we recapitulate the up-to-date knowledge about MRP structure and aligned amino acid sequences to identify the candidate MRP4 residues where cyclic nucleotides bind. We have also listed the most relevant MRP inhibitors studied to date, considering drug safety and specificity for MRP4 in particular. This meta-analysis platform may serve as a basis for the future development of inhibitors of MRP4 cAMP specific transport.


Subject(s)
Cyclic AMP/metabolism , Drug Design , Multidrug Resistance-Associated Proteins/drug effects , Animals , Binding Sites , Humans
11.
Biochem Pharmacol ; 146: 117-126, 2017 12 15.
Article in English | MEDLINE | ID: mdl-28962836

ABSTRACT

Despite the pivotal role GPCRs play in cellular signaling, it is only in the recent years that structural biology has begun to elucidate how GPCRs function and to provide a platform for structure-based drug design. It is postulated that GPCR activation involves the movement of transmembrane helices. The finding that many residues, which have been shown to be critical for receptor activation and are highly conserved among different GPCRs, are clustered in particular positions of transmembrane helices suggests that activation of GPCRs may involve common molecular mechanisms. In particular, phenylalanine 6.44, located in the upper half of TMVI, is highly conserved among almost all GPCRs. We generated Phe 2436.44 Ala/Ser mutants of histamine H2 receptor and found that while the substitutions do not affect receptor expression or ligand signaling, are able to specifically alter cimetidine and ranitidine mechanisms of action from simply inactivating the receptor to produce a ligand-induced G-protein sequestering conformation, that interferes with the signaling of ß2-adrenoceptor. Taking advantage of the cubic ternary complex model, and mathematically modeling our results, we hypothesize that this alteration in ligand mechanism of action is consequence of a change in ligand-induced conformational rearrangement of receptor and its effect on G-protein coupling. Our results show that receptor point mutations can not only alter receptor behavior, as shown for activating/inactivating mutations, but also can have more subtle effects changing ligand mechanism of action.


Subject(s)
Cimetidine/pharmacology , Histamine H2 Antagonists/pharmacology , Ranitidine/pharmacology , Receptors, Histamine H2/genetics , HEK293 Cells , Humans , Models, Molecular , Mutation , Protein Conformation
12.
Handb Exp Pharmacol ; 241: 141-160, 2017.
Article in English | MEDLINE | ID: mdl-27316911

ABSTRACT

Acute myeloid leukemia (AML) consists in a cancer of early hematopoietic cells arising in the bone marrow, most often of those cells that would turn into white blood cells (except lymphocytes). Chemotherapy is the treatment of choice for AML but one of the major complications is that current drugs are highly toxic and poorly tolerated. In general, treatment for AML consists of induction chemotherapy and post-remission therapy. If no further post-remission is given, almost all patients will eventually relapse. Histamine, acting at histamine type-2 (H2) receptors on phagocytes and AML blast cells, helps prevent the production and release of oxygen-free radicals, thereby protecting NK and cytotoxic T cells. This protection allows immune-stimulating agents, such as interleukin-2 (IL-2), to activate cytotoxic cells more effectively, enhancing the killing of tumor cells. Based on this mechanism, post-remission therapy with histamine and IL-2 was found to significantly prevent relapse of AML. Alternatively, another potentially less toxic approach to treat AML employs drugs to induce differentiation of malignant cells. It is based on the assumption that many neoplastic cell types exhibit reversible defects in differentiation, which upon appropriate treatment results in tumor reprogramming and the induction of terminal differentiation. There are promissory results showing that an elevated and sustained signaling through H2 receptors is able to differentiate leukemia-derived cell lines, opening the door for the use of H2 agonists for specific differentiation therapies. In both situations, histamine acting through H2 receptors constitutes an eligible treatment to induce leukemic cell differentiation, improving combined therapies.


Subject(s)
Blood Cells/metabolism , Leukemia, Myeloid, Acute/metabolism , Receptors, Histamine H2/blood , Receptors, Histamine H2/metabolism , Histamine/metabolism , Humans , Interleukin-2/metabolism , Leukemia, Myeloid, Acute/blood , Male
13.
Biochim Biophys Acta ; 1860(9): 1998-2007, 2016 09.
Article in English | MEDLINE | ID: mdl-27316323

ABSTRACT

BACKGROUND: Histamine, through histamine H2 receptor (H2R), modulates different biological processes, involving the modulation of PI3K/AKT/mTOR and RAS/RAF/MEK/ERK pathways. Many evidences have demonstrated the existence and importance of the crossregulation between these two signaling pathways. The aim of the present work was to determine the molecular mechanisms leading to PI3K and ERK pathways modulation induced by the H2R agonist amthamine and to evaluate the possible interplay between them. METHODS: Phosphorylation levels of ERK and Akt were examined by Western blot in HEK293T cells expressing the human H2R, in the presence of H2R agonist and dominant negative mutants or pharmacological inhibitors of different proteins/pathways. Transcriptional activity assays were assessed to determine SRE activity. Amthamine-mediated cellular proliferation was investigated in MA-10A cells in the presence of PI3K inhibitor. RESULTS: H2R agonist inhibits PI3K/Akt/mTOR and stimulates Ras/MEK/ERK pathways. Moreover, PI3K/Akt/mTOR signaling inhibition is necessary to achieve H2R mediated ERK activation. In the presence of a constitutive active mutant of Akt, amthamine is not able to mediate ERK activation. This crosstalk affects classical ERK downstream targets such as Elk1 phosphorylation and the transcriptional activity of the SRE, classically associated to proliferation. We further demonstrate that amthamine-induced proliferation in Leydig MA-10 tumor cells, is enhanced by LY294002, a PI3K inhibitor. CONCLUSIONS: These results describe a crosstalk between PI3K/AKT/mTOR and Ras/MEK/ERK pathways induced by H2R stimulation with implications in cell proliferation. GENERAL SIGNIFICANCE: This work indicates that the modulation of PI3K/AKT/mTOR pathway by H2R in turn regulates Ras/MEK/ERK activation conditioning the proliferative capacity of the cells.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Histamine Agonists/pharmacology , Histamine/metabolism , MAP Kinase Signaling System/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Cell Line , Cell Proliferation/drug effects , HEK293 Cells , Humans , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Thiazoles/pharmacology
14.
Sci Rep ; 5: 17476, 2015 Dec 04.
Article in English | MEDLINE | ID: mdl-26635083

ABSTRACT

Histamine H1 receptor (H1R) antagonists and glucocorticoid receptor (GR) agonists are used to treat inflammatory conditions such as allergic rhinitis, atopic dermatitis and asthma. Consistent with the high morbidity levels of such inflammatory conditions, these receptors are the targets of a vast number of approved drugs, and in many situations their ligands are co-administered. However, this drug association has no clear rationale and has arisen from clinical practice. We hypothesized that H1R signaling could affect GR-mediated activity, impacting on its transcriptional outcome. Indeed, our results show a dual regulation of GR activity by the H1R: a potentiation mediated by G-protein ßγ subunits and a parallel inhibitory effect mediated by Gαq-PLC pathway. Activation of the H1R by its full agonists resulted in a composite potentiating effect. Intriguingly, inactivation of the Gαq-PLC pathway by H1R inverse agonists resulted also in a potentiation of GR activity. Moreover, histamine and clinically relevant antihistamines synergized with the GR agonist dexamethasone to induce gene transactivation and transrepression in a gene-specific manner. Our work provides a delineation of molecular mechanisms underlying the widespread clinical association of antihistamines and GR agonists, which may contribute to future dosage optimization and reduction of well-described side effects associated with glucocorticoid administration.


Subject(s)
Asthma/drug therapy , Dermatitis, Atopic/drug therapy , Receptors, Glucocorticoid/metabolism , Receptors, Histamine H1/metabolism , Asthma/metabolism , Asthma/pathology , Corticosterone/administration & dosage , Dermatitis, Atopic/metabolism , Dermatitis, Atopic/pathology , Dexamethasone/administration & dosage , HeLa Cells , Histamine/metabolism , Histamine Agonists/administration & dosage , Histamine Agonists/metabolism , Histamine H1 Antagonists/metabolism , Humans , Receptors, Glucocorticoid/genetics , Receptors, Histamine H1/genetics , Signal Transduction , Transcriptional Activation/drug effects
15.
PLoS One ; 10(9): e0136878, 2015.
Article in English | MEDLINE | ID: mdl-26360247

ABSTRACT

Anti-mitotic therapies have been considered a hallmark in strategies against abnormally proliferating cells. Focusing on the extensively studied family of thiosemicarbazone (TSC) compounds, we have previously identified 4,4'-dimethoxybenzophenone thiosemicarbazone (T44Bf) as a promising pharmacological compound in a panel of human leukemia cell lines (HL60, U937, KG1a and Jurkat). Present findings indicate that T44Bf-mediated antiproliferative effects are associated with a reversible chronic mitotic arrest caused by defects in chromosome alignment, followed by induced programmed cell death. Furthermore, T44Bf selectively induces apoptosis in leukemia cell lines when compared to normal peripheral blood mononuclear cells. The underlying mechanism of action involves the activation of the mitochondria signaling pathway, with loss of mitochondrial membrane potential and sustained phosphorylation of anti-apoptotic protein Bcl-xL as well as increased Bcl-2 (enhanced phosphorylated fraction) and pro-apoptotic protein Bad levels. In addition, ERK signaling pathway activation was found to be a requisite for T44Bf apoptotic activity. Our findings further describe a novel activity for a benzophenone thiosemicarbazone and propose T44Bf as a promising anti-mitotic prototype to develop chemotherapeutic agents to treat acute leukemia malignancies.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Thiosemicarbazones/pharmacology , Antineoplastic Agents/chemistry , Benzophenones/chemistry , Biomarkers , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin A1/genetics , Cyclin A1/metabolism , Cyclin B1/genetics , Cyclin B1/metabolism , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mitosis/drug effects , Mitosis/genetics , Signal Transduction/drug effects , Thiosemicarbazones/chemistry
16.
PLoS One ; 10(3): e0120651, 2015.
Article in English | MEDLINE | ID: mdl-25790437

ABSTRACT

Cyclic AMP represents one of the most studied signaling molecules and its role in proliferation and differentiation processes has been well established. Intracellular cAMP levels are tightly regulated where the MRP4 transporter plays a major role. In the present study, we sought to establish whether cAMP modulated MRP4 expression in pancreatic adenocarcinoma cell lines. Quantitative PCR and western blot studies showed that cAMP-increasing agents enhanced MRP4 transcripts and protein levels in PANC-1 cells. Reporter luciferase experiments carried out in pancreatic AR42J cells showed that intracellular cAMP up-regulates MRP4 through an Epac2- and Rap1-mediated mechanism whereas extracellular cAMP reduced MRP4 promoter activity by a MEK/ERK-mediated pathway. Present results show that cAMP regulates MRP4 promoter activity, and further indicate that the balance between intracellular and extracellular cAMP levels determines MRP4 expression.


Subject(s)
Adenocarcinoma/genetics , Cyclic AMP/metabolism , Gene Expression Regulation, Neoplastic , Multidrug Resistance-Associated Proteins/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Transcription, Genetic , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Humans , Pancreatic Neoplasms/pathology , Promoter Regions, Genetic/genetics , Signal Transduction
17.
Front Pharmacol ; 6: 45, 2015.
Article in English | MEDLINE | ID: mdl-25805997

ABSTRACT

Histamine mediates numerous functions acting through its four receptor subtypes all belonging to the large family of seven transmembrane G-protein coupled receptors. In particular, histamine H2 receptor (H2R) is mainly involved in gastric acid production, becoming a classic pharmacological target to treat Zollinger-Ellison disease and gastric and duodenal ulcers. H2 ligands rank among the most widely prescribed and over the counter-sold drugs in the world. Recent evidence indicate that some H2R ligands display biased agonism, selecting and triggering some, but not all, of the signaling pathways associated to the H2R. The aim of the present work is to study whether famotidine, clinically widespread used ligand acting at H2R, exerts biased signaling. Our findings indicate that while famotidine acts as inverse agonist diminishing cAMP basal levels, it mimics the effects of histamine and the agonist amthamine concerning receptor desensitization and internalization. Moreover, the treatment of HEK293T transfected cells with any of the three ligands lead to a concentration dependent pERK increment. Similarly in AGS gastric epithelial cells, famotidine treatment led to both, the reduction in cAMP levels as well as the increment in ERK phosphorylation, suggesting that this behavior could have pharmacological relevant implications. Based on that, histidine decarboxylase expression was studied by quantitative PCR in AGS cells and its levels were increased by famotidine as well as by histamine and amthamine. In all cases, the positive regulation was impeded by the MEK inhibitor PD98059, indicating that biased signaling toward ERK1/2 pathway is the responsible of such enzyme regulation. These results support that ligand bias is not only a pharmacological curiosity but has physiological and pharmacological implications on cell metabolism.

18.
Oncotarget ; 5(19): 9308-21, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25301721

ABSTRACT

Less than a third of adults patients with acute myeloid leukemia (AML) are cured by current treatments, emphasizing the need for new approaches to therapy. We previously demonstrated that besides playing a role in drug-resistant leukemia cell lines, multidrug resistance protein 4 (MRP4/ABCC4) regulates leukemia cell proliferation and differentiation through the endogenous MRP4/ABCC4 substrate, cAMP. Here, we studied the role of MRP4/ABCC4 in tumor progression in a mouse xenograft model and in leukemic stem cells (LSCs) differentiation. We found a decrease in the mitotic index and an increase in the apoptotic index associated with the inhibition of tumor growth when mice were treated with rolipram (PDE4 inhibitor) and/or probenecid (MRPs inhibitor). Genetic silencing and pharmacologic inhibition of MRP4 reduced tumor growth. Furthermore, MRP4 knockdown induced cell cycle arrest and apoptosis in vivo. Interestingly, when LSC population was isolated, we observed that increased cAMP levels and MRP4/ABCC4 blockade resulted in LSCs differentiation. Taken together, our findings show that MRP4/ABCC4 has a relevant role in tumor growth and apoptosis and in the eradication of LSCs, providing the basis for a novel promising target in AML therapy.


Subject(s)
Apoptosis/genetics , Cell Cycle Checkpoints/genetics , Leukemia, Myeloid, Acute/pathology , Multidrug Resistance-Associated Proteins/genetics , Neoplastic Stem Cells/cytology , Animals , Apoptosis/drug effects , Cell Differentiation , Cell Line, Tumor , Cell Proliferation/genetics , Cyclic AMP/metabolism , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Disease Progression , Female , Humans , Leukemia, Myeloid, Acute/drug therapy , Mice , Mice, Nude , Mitotic Index , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Neoplasm Transplantation , Phosphodiesterase 4 Inhibitors/pharmacology , RNA Interference , RNA, Small Interfering/biosynthesis , RNA, Small Interfering/genetics , Rolipram/pharmacology , Transplantation, Heterologous
19.
Biochem J ; 459(1): 117-26, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24417223

ABSTRACT

7TMRs (seven-transmembrane receptors) exist as conformational collections in which different conformations would lead to differential downstream behaviours such as receptor phosphorylation, G-protein activation and receptor internalization. In this context, a ligand may cause differential activation of some, but not all, of the signalling events, which are associated to a particular receptor, and it would lead to biased agonism. The aim of the present study was to investigate whether H2R (histamine H2 receptor) ligands, described as inverse agonists because of their negative efficacy at modulating adenylate cyclase, could display some positive efficacy concerning receptor desensitization, internalization or even signalling through an adenylate-cyclase-independent pathway. Our present findings indicate that treatment with H2R inverse agonists leads to receptor internalization in HEK (human embryonic kidney)-293T transfected cells, by a mechanism mediated by arrestin and dynamin, but independent of GRK2 (G-protein-coupled receptor kinase 2)-mediated phosphorylation. On the other hand, we prove that two of the H2R inverse agonists tested, ranitidine and tiotidine, also induce receptor desensitization. Finally, we show that these ligands are able to display positive efficacy towards the ERK1/2 (extracellular-signal-regulated kinase 1/2) pathway by a mechanism that involves Gßγ and PI3K (phosphoinositide 3-kinase)-mediated signalling in both transfected HEK-293T cells and human gastric adenocarcinoma cells. These results point to the aspect of pluridimensional efficacy at H2R as a phenomenon that could be extended to naïve cells, and challenge previous classification of pharmacologically relevant histaminergic ligands.


Subject(s)
Drug Inverse Agonism , Histamine H2 Antagonists/metabolism , Receptors, Histamine H2/metabolism , Signal Transduction/physiology , Cell Line, Tumor , HEK293 Cells , Histamine H2 Antagonists/pharmacology , Humans , Ligands , Ranitidine/metabolism , Ranitidine/pharmacology , Signal Transduction/drug effects
20.
Eur J Pharmacol ; 715(1-3): 26-32, 2013 Sep 05.
Article in English | MEDLINE | ID: mdl-23831018

ABSTRACT

The accurate characterization of the molecular mechanisms involved in the action of receptor ligands is important for their appropriate therapeutic use and safety. It is well established that ligands acting at the histamine system currently used in the clinic exert their actions by specifically antagonizing G-protein coupled H1 and H2 receptors. However, most of these ligands, assumed to be neutral antagonists, behave as inverse agonists displaying negative efficacy in experimental systems. This suggests that their therapeutic actions may involve not only receptor blockade, but also the decrease of spontaneous receptor activity. The mechanisms whereby inverse agonists achieve negative efficacy are diverse. Theoretical models predict at least three possible mechanisms, all of which are supported by experimental observations. Depending on the mechanism of action engaged, the inverse agonist could interfere specifically with signaling events triggered by unrelated receptors. This possibility opens up new venues to explain the therapeutic actions of inverse agonists of the histamine receptor and perhaps new therapeutic applications.


Subject(s)
Drug Inverse Agonism , Histamine Antagonists/pharmacology , Animals , Histamine Antagonists/therapeutic use , Humans , Ligands , Models, Molecular , Receptors, Histamine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...