1
|
Sahores A, González AR, Yaneff A, May M, Gómez N, Monczor F, Fernández N, Davio C, Shayo C. Ceefourin-1, a MRP4/ABCC4 inhibitor, induces apoptosis in AML cells enhanced by histamine. Biochim Biophys Acta Gen Subj 2023; 1867:130322. [PMID: 36773726 DOI: 10.1016/j.bbagen.2023.130322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/27/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023]
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.
Collapse
Affiliation(s)
- Ana Sahores
- Laboratorio de Patología y Farmacología Molecular, Instituto de Biología y Medicina Experimental - Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Buenos Aires, Argentina; Instituto de Investigaciones Farmacológicas, ININFA - Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET. Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Angela Rodríguez González
- Laboratorio de Patología y Farmacología Molecular, Instituto de Biología y Medicina Experimental - Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Buenos Aires, Argentina
| | - Agustín Yaneff
- Instituto de Investigaciones Farmacológicas, ININFA - Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET. Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María May
- Instituto de Investigaciones Farmacológicas, ININFA - Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET. Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Natalia Gómez
- Instituto de Investigaciones Farmacológicas, ININFA - Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET. Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Federico Monczor
- Instituto de Investigaciones Farmacológicas, ININFA - Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET. Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Natalia Fernández
- Instituto de Investigaciones Farmacológicas, ININFA - Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET. Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carlos Davio
- Instituto de Investigaciones Farmacológicas, ININFA - Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET. Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carina Shayo
- Laboratorio de Patología y Farmacología Molecular, Instituto de Biología y Medicina Experimental - Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Buenos Aires, Argentina.
| |
Collapse
|
2
|
Colavita JPM, Todaro JS, de Sousa M, May M, Gómez N, Yaneff A, Di Siervi N, Aguirre MV, Guijas C, Ferrini L, Davio C, Rodríguez JP. Multidrug resistance protein 4 (MRP4/ABCC4) is overexpressed in clear cell renal cell carcinoma (ccRCC) and is essential to regulate cell proliferation. Int J Biol Macromol 2020; 161:836-847. [PMID: 32553977 DOI: 10.1016/j.ijbiomac.2020.06.106] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 12/13/2022]
Abstract
Kidney cancer accounts for 2.5% of all cancers, with an annual global incidence of almost 300,000 cases leading to 111,000 deaths. Approximately 85% of kidney tumors are renal cell carcinoma (RCC) and their major histologic subtype is clear cell renal cell carcinoma (ccRCC). Although new therapeutic treatments are being designed and applied based on the combination of tyrosine kinase inhibitors and immunotherapy, no major impact on the mortality has been reported so far. MRP4 is a pump efflux that transporters multiple endogenous and exogenous substances. Recently it has been associated with tumoral persistence and cell proliferation in several types of cancer including pancreas, lung, ovary, colon, ostesarcoma, etc. Herein, we demonstrate for the first time, that MRP4 is overexpressed in ccRCC tumors, compared to control renal tissues. In addition, using cell culture models, we observed that MRP4 pharmacological inhibition produces an imbalance in cAMP metabolism, induces cell arrest, changes in lipid composition, increase in cytoplasmic lipid droplets and finally apoptosis. These data provide solid evidence for the future evaluation of MRP4 as a possible new therapeutic target in ccRCC.
Collapse
Affiliation(s)
- Juan Pablo Melana Colavita
- Laboratorio de Investigaciones Bioquímicas de la Facultad de Medicina (LIBIM), Instituto de Química Básica y Aplicada del NEA, (IQUIBA NEA-UNNE-CONICET), Facultad de Medicina, Universidad Nacional del Nordeste, 3400 Corrientes, Argentina
| | - Juan Santiago Todaro
- Laboratorio de Investigaciones Bioquímicas de la Facultad de Medicina (LIBIM), Instituto de Química Básica y Aplicada del NEA, (IQUIBA NEA-UNNE-CONICET), Facultad de Medicina, Universidad Nacional del Nordeste, 3400 Corrientes, Argentina
| | - Maximiliano de Sousa
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, 1000 Buenos Aires, Argentina
| | - María May
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, 1000 Buenos Aires, Argentina
| | - Natalia Gómez
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, 1000 Buenos Aires, Argentina
| | - Agustin Yaneff
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, 1000 Buenos Aires, Argentina
| | - Nicolas Di Siervi
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, 1000 Buenos Aires, Argentina
| | - María Victoria Aguirre
- Laboratorio de Investigaciones Bioquímicas de la Facultad de Medicina (LIBIM), Instituto de Química Básica y Aplicada del NEA, (IQUIBA NEA-UNNE-CONICET), Facultad de Medicina, Universidad Nacional del Nordeste, 3400 Corrientes, Argentina
| | - Carlos Guijas
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas (CSIC), Universidad de Valladolid, 47003 Valladolid, Spain
| | - Leandro Ferrini
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, 1000 Buenos Aires, Argentina
| | - Carlos Davio
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, 1000 Buenos Aires, Argentina
| | - Juan Pablo Rodríguez
- Laboratorio de Investigaciones Bioquímicas de la Facultad de Medicina (LIBIM), Instituto de Química Básica y Aplicada del NEA, (IQUIBA NEA-UNNE-CONICET), Facultad de Medicina, Universidad Nacional del Nordeste, 3400 Corrientes, Argentina.
| |
Collapse
|
3
|
Rodríguez González A, Sahores A, Díaz-Nebreda A, Yaneff A, Di Siervi N, Gómez N, Monczor F, Fernández N, Davio C, Shayo C. MRP4/ABCC4 expression is regulated by histamine in acute myeloid leukemia cells, determining cAMP efflux. FEBS J 2020; 288:229-243. [PMID: 32333821 DOI: 10.1111/febs.15344] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 03/02/2020] [Accepted: 04/22/2020] [Indexed: 12/14/2022]
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.
Collapse
Affiliation(s)
| | - Ana Sahores
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina
| | - Antonela Díaz-Nebreda
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Agustín Yaneff
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina
| | - Nicolás Di Siervi
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina
| | - Natalia Gómez
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina
| | - Federico Monczor
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina
| | - Natalia Fernández
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina
| | - Carlos Davio
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina
| | - Carina Shayo
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| |
Collapse
|
4
|
Perez DR, Sklar LA, Chigaev A, Matlawska-Wasowska K. Drug repurposing for targeting cyclic nucleotide transporters in acute leukemias - A missed opportunity. Semin Cancer Biol 2020; 68:199-208. [PMID: 32044470 DOI: 10.1016/j.semcancer.2020.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/01/2019] [Accepted: 02/03/2020] [Indexed: 02/08/2023]
Abstract
While current treatment regimens for acute leukemia can dramatically improve patient survival, there remains room for improvement. Due to its roles in cell differentiation, cell survival, and apoptotic signaling, modulation of the cyclic AMP (cAMP) pathway has provided a meaningful target in hematological malignancies. Several studies have demonstrated that gene expression profiles associated with increased pro-survival cAMP activity or downregulation of various pro-apoptotic factors associated with the cAMP pathway are apparent in acute leukemia patients. Previous work to increase leukemia cell intracellular cAMP focused on the use of cAMP analogs, stimulating cAMP production via transmembrane-associated adenylyl cyclases, or decreasing cAMP degradation by inhibiting phosphodiesterase activity. However, targeting cyclic nucleotide efflux by ATP-binding cassette (ABC) transporters represents an unexplored approach for modulation of intracellular cyclic nucleotide levels. Preliminary studies have shown that inhibition of cAMP efflux can stimulate leukemia cell differentiation, cell growth arrest, and apoptosis, indicating that targeting cAMP efflux may show promise for future therapeutic development. Furthermore, inhibition of cyclic nucleotide transporter activity may also contribute multiple anticancer benefits by reducing extracellular pro-survival signaling in malignant cells. Hence, several opportunities for drug repurposing may exist for targeting cyclic nucleotide transporters.
Collapse
Affiliation(s)
- Dominique R Perez
- Department of Pathology, Health Sciences Center, University of New Mexico, Albuquerque, NM, USA; Center for Molecular Discovery, Health Sciences Center, University of New Mexico, Albuquerque, NM, USA
| | - Larry A Sklar
- Department of Pathology, Health Sciences Center, University of New Mexico, Albuquerque, NM, USA; Center for Molecular Discovery, Health Sciences Center, University of New Mexico, Albuquerque, NM, USA; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
| | - Alexandre Chigaev
- Department of Pathology, Health Sciences Center, University of New Mexico, Albuquerque, NM, USA; Center for Molecular Discovery, Health Sciences Center, University of New Mexico, Albuquerque, NM, USA; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA.
| | - Ksenia Matlawska-Wasowska
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA; Department of Pediatrics, Division of Hematology-Oncology, Health Sciences Center, University of New Mexico, Albuquerque, NM, USA.
| |
Collapse
|
5
|
Abreu M, Basti A, Genov N, Mazzoccoli G, Relógio A. The reciprocal interplay between TNFα and the circadian clock impacts on cell proliferation and migration in Hodgkin lymphoma cells. Sci Rep 2018; 8:11474. [PMID: 30065253 PMCID: PMC6068144 DOI: 10.1038/s41598-018-29847-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 07/18/2018] [Indexed: 12/20/2022] Open
Abstract
A bidirectional interaction between the circadian network and effector mechanisms of immunity brings on a proper working of both systems. In the present study, we used Hodgkin lymphoma (HL) as an experimental model for a type of cancer involving cells of the immune system. We identified this cancer type among haematological malignancies has having a strong differential expression of core-clock elements. Taking advantage of bioinformatics analyses and experimental procedures carried out in III- and IV-stage HL cells, and lymphoblastoid B cells, we explored this interplay and bear out diverse interacting partners of both systems. In particular, we assembled a wide-ranging network of clock-immune-related genes and pinpointed TNF as a crucial intermediary player. A robust circadian clock hallmarked III-stage lymphoma cells, differently from IV-stage HL cells, which do not harbour a properly functioning clockwork. TNF and circadian gene modulation impacted on clock genes expression and triggered phenotypic changes in lymphoma cells, suggesting a crucial involvement of core-clock elements and TNF in the physiopathological mechanisms hastening malignancy. Our results move forward our understanding of the putative role of the core-clock and TNF in the pathobiology of Hodgkin lymphoma, and highlight their influence in cellular proliferation and migration in lymphatic cancers.
Collapse
Affiliation(s)
- Mónica Abreu
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt - Universität zu Berlin, and Berlin Institute of Health, Institute for Theoretical Biology, Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt - Universität zu Berlin, and Berlin Institute of Health, Medical Department of Hematology, Oncology, and Tumor Immunology, Molecular Cancer Research Center, Berlin, Germany
| | - Alireza Basti
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt - Universität zu Berlin, and Berlin Institute of Health, Institute for Theoretical Biology, Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt - Universität zu Berlin, and Berlin Institute of Health, Medical Department of Hematology, Oncology, and Tumor Immunology, Molecular Cancer Research Center, Berlin, Germany
| | - Nikolai Genov
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt - Universität zu Berlin, and Berlin Institute of Health, Institute for Theoretical Biology, Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt - Universität zu Berlin, and Berlin Institute of Health, Medical Department of Hematology, Oncology, and Tumor Immunology, Molecular Cancer Research Center, Berlin, Germany
| | - Gianluigi Mazzoccoli
- Department of Medical Sciences, Division of Internal Medicine and Chronobiology Unit, IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo (FG), Italy
| | - Angela Relógio
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt - Universität zu Berlin, and Berlin Institute of Health, Institute for Theoretical Biology, Berlin, Germany. .,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt - Universität zu Berlin, and Berlin Institute of Health, Medical Department of Hematology, Oncology, and Tumor Immunology, Molecular Cancer Research Center, Berlin, Germany.
| |
Collapse
|
6
|
Illiano M, Conte M, Sapio L, Nebbioso A, Spina A, Altucci L, Naviglio S. Forskolin Sensitizes Human Acute Myeloid Leukemia Cells to H3K27me2/3 Demethylases GSKJ4 Inhibitor via Protein Kinase A. Front Pharmacol 2018; 9:792. [PMID: 30079022 PMCID: PMC6063003 DOI: 10.3389/fphar.2018.00792] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/28/2018] [Indexed: 01/26/2023] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive hematological malignancy occurring very often in older adults, with poor prognosis depending on both rapid disease progression and drug resistance occurrence. Therefore, new therapeutic approaches are demanded. Epigenetic marks play a relevant role in AML. GSKJ4 is a novel inhibitor of the histone demethylases JMJD3 and UTX. To note GSKJ4 has been recently shown to act as a potent small molecule inhibitor of the proliferation in many cancer cell types. On the other hand, forskolin, a natural cAMP raising compound, used for a long time in traditional medicine and considered safe also in recent studies, is emerging as a very interesting molecule for possible use in cancer therapy. Here, we investigate the effects of forskolin on the sensitivity of human leukemia U937 cells to GSKJ4 through flow cytometry-based assays (cell-cycle progression and cell death), cell number counting, and immunoblotting experiments. We provide evidence that forskolin markedly potentiates GSKJ4-induced antiproliferative effects by apoptotic cell death induction, accompanied by a dramatic BCL2 protein down-regulation as well as caspase 3 activation and PARP protein cleavage. Comparable effects are observed with the phosphodiesterase inhibitor IBMX and 8-Br-cAMP analogous, but not by using 8-pCPT-2'-O-Me-cAMP Epac activator. Moreover, the forskolin-induced enhancement of sensitivity to GSKJ4 is counteracted by pre-treatment with Protein Kinase A (PKA) inhibitors. Altogether, our data strongly suggest that forskolin sensitizes U937 cells to GSKJ4 inhibitor via a cAMP/PKA-mediated mechanism. Our findings provide initial evidence of anticancer activity induced by forskolin/GSKJ4 combination in leukemia cells and underline the potential for use of forskolin and GSKJ4 in the development of innovative and effective therapeutic approaches for AML treatment.
Collapse
Affiliation(s)
- Michela Illiano
- Department of Precision Medicine, School of Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | | | - Luigi Sapio
- Department of Precision Medicine, School of Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Angela Nebbioso
- Department of Precision Medicine, School of Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Annamaria Spina
- Department of Precision Medicine, School of Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Lucia Altucci
- Department of Precision Medicine, School of Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Silvio Naviglio
- Department of Precision Medicine, School of Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
7
|
Monczor F, Copsel S, Fernandez N, Davio C, Shayo C. Histamine H 2 Receptor in Blood Cells: A Suitable Target for the Treatment of Acute Myeloid Leukemia. Handb Exp Pharmacol 2017; 241:141-160. [PMID: 27316911 DOI: 10.1007/164_2016_8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
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.
Collapse
Affiliation(s)
- Federico Monczor
- Instituto de Investigaciones Farmacológicas, ININFA, Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Junín 956 PP, (1113), Buenos Aires, Argentina.
| | - Sabrina Copsel
- Microbiology and Immunology Department, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Natalia Fernandez
- Instituto de Investigaciones Farmacológicas, ININFA, Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Junín 956 PP, (1113), Buenos Aires, Argentina
| | - Carlos Davio
- Instituto de Investigaciones Farmacológicas, ININFA, Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Junín 956 PP, (1113), Buenos Aires, Argentina
| | - Carina Shayo
- Laboratorio de Patología y Farmacología Molecular, Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| |
Collapse
|
8
|
Wen J, Luo J, Huang W, Tang J, Zhou H, Zhang W. The Pharmacological and Physiological Role of Multidrug-Resistant Protein 4. J Pharmacol Exp Ther 2015; 354:358-75. [PMID: 26148856 DOI: 10.1124/jpet.115.225656] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Accepted: 06/30/2015] [Indexed: 12/11/2022] Open
Abstract
Multidrug-resistant protein 4 (MRP4), a member of the C subfamily of ATP-binding cassette transporters, is distributed in a variety of tissues and a number of cancers. As a drug transporter, MRP4 is responsible for the pharmacokinetics and pharmacodynamics of numerous drugs, especially antiviral drugs, antitumor drugs, and diuretics. In this regard, the functional role of MRP4 is affected by a number of factors, such as genetic mutations; tissue-specific transcriptional regulations; post-transcriptional regulations, including miRNAs and membrane internalization; and substrate competition. Unlike other C family members, MRP4 is in a pivotal position to transport cellular signaling molecules, through which it is tightly connected to the living activity and physiologic processes of cells and bodies. In the context of several cancers in which MRP4 is overexpressed, MRP4 inhibition shows striking effects against cancer progression and drug resistance. In this review, we describe the role of MRP4 more specifically in both healthy conditions and disease states, with an emphasis on its potential as a drug target.
Collapse
Affiliation(s)
- Jiagen Wen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, ChangSha, P.R. China; Institute of Clinical Pharmacology, Central South University, ChangSha, P.R. China; and Hunan Key Laboratory of Pharmacogenetics, ChangSha, P.R. China
| | - Jianquan Luo
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, ChangSha, P.R. China; Institute of Clinical Pharmacology, Central South University, ChangSha, P.R. China; and Hunan Key Laboratory of Pharmacogenetics, ChangSha, P.R. China
| | - Weihua Huang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, ChangSha, P.R. China; Institute of Clinical Pharmacology, Central South University, ChangSha, P.R. China; and Hunan Key Laboratory of Pharmacogenetics, ChangSha, P.R. China
| | - Jie Tang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, ChangSha, P.R. China; Institute of Clinical Pharmacology, Central South University, ChangSha, P.R. China; and Hunan Key Laboratory of Pharmacogenetics, ChangSha, P.R. China
| | - Honghao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, ChangSha, P.R. China; Institute of Clinical Pharmacology, Central South University, ChangSha, P.R. China; and Hunan Key Laboratory of Pharmacogenetics, ChangSha, P.R. China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, ChangSha, P.R. China; Institute of Clinical Pharmacology, Central South University, ChangSha, P.R. China; and Hunan Key Laboratory of Pharmacogenetics, ChangSha, P.R. China
| |
Collapse
|
9
|
The role of cyclic nucleotide signaling pathways in cancer: targets for prevention and treatment. Cancers (Basel) 2014; 6:436-58. [PMID: 24577242 PMCID: PMC3980602 DOI: 10.3390/cancers6010436] [Citation(s) in RCA: 145] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 01/10/2014] [Accepted: 02/07/2014] [Indexed: 12/13/2022] Open
Abstract
For more than four decades, the cyclic nucleotides cyclic AMP (cAMP) and cyclic GMP (cGMP) have been recognized as important signaling molecules within cells. Under normal physiological conditions, cyclic nucleotides regulate a myriad of biological processes such as cell growth and adhesion, energy homeostasis, neuronal signaling, and muscle relaxation. In addition, altered cyclic nucleotide signaling has been observed in a number of pathophysiological conditions, including cancer. While the distinct molecular alterations responsible for these effects vary depending on the specific cancer type, several studies have demonstrated that activation of cyclic nucleotide signaling through one of three mechanisms-induction of cyclic nucleotide synthesis, inhibition of cyclic nucleotide degradation, or activation of cyclic nucleotide receptors-is sufficient to inhibit proliferation and activate apoptosis in many types of cancer cells. These findings suggest that targeting cyclic nucleotide signaling can provide a strategy for the discovery of novel agents for the prevention and/or treatment of selected cancers.
Collapse
|
10
|
Pagotto RM, Monzón C, Moreno MB, Pignataro OP, Mondillo C. Proliferative effect of histamine on MA-10 Leydig tumor cells mediated through HRH2 activation, transient elevation in cAMP production, and increased extracellular signal-regulated kinase phosphorylation levels. Biol Reprod 2012; 87:150. [PMID: 23077168 DOI: 10.1095/biolreprod.112.102905] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Mast cells (MC) occur normally in the testis with a species-specific distribution, yet their precise role remains unclear. Testicular MC express histidine decarboxylase (HDC), the unique enzyme responsible for histamine (HA) generation. Evidence to date supports a role for HA as a local regulator of steroidogenesis via functional H₁ and H₂ receptor subtypes (HRH1 and HRH2, respectively) present in Leydig cells. Given that HA is a well-known modulator of physiological and pathological proliferation in many different cell types, we aimed in the present study to evaluate whether HA might contribute to the regulation of Leydig cell number as well as to the control of androgen production. Herein, we demonstrate, to our knowledge for the first time, that MA-10 Leydig tumor cells, but not normal immature Leydig cells (ILC), exhibit a proliferative response upon stimulation with HA that involves HRH2 activation, transient elevation of cAMP levels, and increased extracellular signal-regulated kinase (ERK) phosphorylation. Our results also reveal that MA-10 cells show significantly heightened HDC expression compared to normal ILC or whole-testicular lysate and that inhibition of HDC activity decreases MA-10 cell proliferation, suggesting a possible correlation between autocrine overproduction of HA and abnormally increased proliferation in Leydig cells. The facts that germ cells are also both source and target of HA and that multiple testicular cells are susceptible to HA action underline the importance of the present study, which we hope will serve as a first step for further research into regulation of non-MC-related HDC expression within the testis and its significance for testicular function.
Collapse
Affiliation(s)
- Romina María Pagotto
- Laboratory of Molecular Endocrinology and Signal Transduction, Institute of Biology and Experimental Medicine, National Research Council (IByME-CONICET), Buenos Aires, Argentina
| | | | | | | | | |
Collapse
|
11
|
Hertz AL, Beavo JA. Cyclic nucleotides and phosphodiesterases in monocytic differentiation. Handb Exp Pharmacol 2011:365-90. [PMID: 21695649 DOI: 10.1007/978-3-642-17969-3_16] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Monocytes are immune cells that can differentiate into a number of cell types including macrophages, dendritic cells, and osteoclasts upon exposure to various cytokines. The phenotypes of these differentiated cells are highly heterogeneous and their differentiation can be affected by the cyclic nucleotides, 3'-5'-cyclic adenosine monophosphate (cAMP) and 3'-5'-cyclic guanosine monophosphate (cGMP). The intracellular levels of cAMP and cGMP are controlled through regulation of production by adenylyl and guanylyl cyclases and through degradation by cyclic nucleotide phosphodiesterases (PDEs). PDE inhibition and subsequent changes in cyclic nucleotide levels can alter the final phenotype of a differentiating monocyte with regards to surface marker expression, gene expression, or changes in secreted chemokine and cytokine levels. The differentiation process itself can also be either inhibited or augmented by changes in cyclic nucleotide levels, depending on the system being studied and the timing of cyclic nucleotide elevation. This chapter explores the effects of PDE inhibition and increases in cGMP and cAMP on monocytic differentiation into osteoclasts, dendritic cells, and macrophages.
Collapse
Affiliation(s)
- Angie L Hertz
- Department of Pharmacology, School of Medicine, University of Washington, 357280, Seattle, WA 98125, USA.
| | | |
Collapse
|
12
|
Fernandez N, Gottardo FL, Alonso MN, Monczor F, Shayo C, Davio C. Roles of phosphorylation-dependent and -independent mechanisms in the regulation of histamine H2 receptor by G protein-coupled receptor kinase 2. J Biol Chem 2011; 286:28697-28706. [PMID: 21705320 DOI: 10.1074/jbc.m111.269613] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
It is widely assumed that G protein-coupled receptor kinase 2 (GRK2)-mediated specific inhibition of G protein-coupled receptors (GPCRs) response involves GRK-mediated receptor phosphorylation followed by β-arrestin binding and subsequent uncoupling from the heterotrimeric G protein. It has recently become evident that GRK2-mediated GPCRs regulation also involves phosphorylation-independent mechanisms. In the present study we investigated whether the histamine H2 receptor (H2R), a Gα(s)-coupled GPCR known to be desensitized by GRK2, needs to be phosphorylated for its desensitization and/or internalization and resensitization. For this purpose we evaluated the effect of the phosphorylating-deficient GRK2K220R mutant on H2R signaling in U937, COS7, and HEK293T cells. We found that although this mutant functioned as dominant negative concerning receptor internalization and resensitization, it desensitized H2R signaling in the same degree as the GRK2 wild type. To identify the domains responsible for the kinase-independent receptor desensitization, we co-transfected the receptor with constructions encoding the GRK2 RGS-homology domain (RH) and the RH or the kinase domain fused to the pleckstrin-homology domain. Results demonstrated that the RH domain of GRK2 was sufficient to desensitize the H2R. Moreover, disruption of RGS functions by the use of GRK2D110A/K220R double mutant, although coimmunoprecipitating with the H2R, reversed GRK2K220R-mediated H2R desensitization. Overall, these results indicate that GRK2 induces desensitization of H2R through a phosphorylation-independent and RGS-dependent mechanism and extends the GRK2 RH domain-mediated regulation of GPCRs beyond Gα(q)-coupled receptors. On the other hand, GRK2 kinase activity proved to be necessary for receptor internalization and the resulting resensitization.
Collapse
Affiliation(s)
- Natalia Fernandez
- Laboratorio de Farmacología de Receptores, Cátedra de Química Medicinal, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, 1113 Buenos Aires, Argentina,; Consejo Nacional de Investigaciones Científicas y Técnicas, 1033 Buenos Aires, Argentina.
| | - Federico L Gottardo
- Laboratorio de Farmacología de Receptores, Cátedra de Química Medicinal, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, 1113 Buenos Aires, Argentina
| | - Maria N Alonso
- Consejo Nacional de Investigaciones Científicas y Técnicas, 1033 Buenos Aires, Argentina; Laboratorio de Farmacología y Patología Molecular, Instituto de Biología y Medicina Experimental, 1426 Buenos Aires, Argentina, and
| | - Federico Monczor
- Laboratorio de Farmacología de Receptores, Cátedra de Química Medicinal, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, 1113 Buenos Aires, Argentina,; Consejo Nacional de Investigaciones Científicas y Técnicas, 1033 Buenos Aires, Argentina
| | - Carina Shayo
- Consejo Nacional de Investigaciones Científicas y Técnicas, 1033 Buenos Aires, Argentina; Laboratorio de Farmacología y Patología Molecular, Instituto de Biología y Medicina Experimental, 1426 Buenos Aires, Argentina, and
| | - Carlos Davio
- Laboratorio de Farmacología de Receptores, Cátedra de Química Medicinal, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, 1113 Buenos Aires, Argentina,; Consejo Nacional de Investigaciones Científicas y Técnicas, 1033 Buenos Aires, Argentina
| |
Collapse
|
13
|
Copsel S, Garcia C, Diez F, Vermeulem M, Baldi A, Bianciotti LG, Russel FGM, Shayo C, Davio C. Multidrug resistance protein 4 (MRP4/ABCC4) regulates cAMP cellular levels and controls human leukemia cell proliferation and differentiation. J Biol Chem 2011; 286:6979-88. [PMID: 21205825 PMCID: PMC3044954 DOI: 10.1074/jbc.m110.166868] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Revised: 01/03/2011] [Indexed: 11/06/2022] Open
Abstract
Increased intracellular cAMP concentration plays a well established role in leukemic cell maturation. We previously reported that U937 cells stimulated by H2 receptor agonists, despite a robust increase in cAMP, fail to mature because of rapid H2 receptor desensitization and phosphodiesterase (PDE) activation. Here we show that intracellular cAMP levels not only in U937 cells but also in other acute myeloid leukemia cell lines are also regulated by multidrug resistance-associated proteins (MRPs), particularly MRP4. U937, HL-60, and KG-1a cells, exposed to amthamine (H2-receptor agonist), augmented intracellular cAMP concentration with a concomitant increase in the efflux. Extrusion of cAMP was ATP-dependent and probenecid-sensitive, supporting that the transport was MRP-mediated. Cells exposed to amthamine and the PDE4 inhibitor showed enhanced cAMP extrusion, but this response was inhibited by MRP blockade. Amthamine stimulation, combined with PDE4 and MRP inhibition, induced maximal cell arrest proliferation. Knockdown strategy by shRNA revealed that this process was mediated by MRP4. Furthermore, blockade by probenecid or MRP4 knockdown showed that increased intracellular cAMP levels induce maturation in U937 cells. These findings confirm the key role of intracellular cAMP levels in leukemic cell maturation and provide the first evidence that MRP4 may represent a new potential target for leukemia differentiation therapy.
Collapse
Affiliation(s)
- Sabrina Copsel
- From the Laboratorio de Farmacología de Receptores, Cátedra de Química Medicinal, Departamento de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, 1113 Buenos Aires, Argentina
- the Laboratorio de Patología y Farmacología Molecular, Instituto de Biología y Medicina Experimental, 1428 Buenos Aires, Argentina
- the Consejo Nacional de Investigaciones Científicas y Técnicas and
| | - Corina Garcia
- From the Laboratorio de Farmacología de Receptores, Cátedra de Química Medicinal, Departamento de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, 1113 Buenos Aires, Argentina
- the Consejo Nacional de Investigaciones Científicas y Técnicas and
| | - Federico Diez
- From the Laboratorio de Farmacología de Receptores, Cátedra de Química Medicinal, Departamento de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, 1113 Buenos Aires, Argentina
| | - Monica Vermeulem
- the Consejo Nacional de Investigaciones Científicas y Técnicas and
- Departamento de Inmunología, Instituto de Investigaciones Hematológicas, Academia Nacional de Medicina, 1425 Buenos Aires, Argentina
| | - Alberto Baldi
- the Laboratorio de Patología y Farmacología Molecular, Instituto de Biología y Medicina Experimental, 1428 Buenos Aires, Argentina
- the Consejo Nacional de Investigaciones Científicas y Técnicas and
| | - Liliana G. Bianciotti
- the Consejo Nacional de Investigaciones Científicas y Técnicas and
- the Cátedra de Fisiopatología, Departamento de Ciencias Biológicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, 1113 Buenos Aires, Argentina, and
| | - Frans G. M. Russel
- the Department of Pharmacology and Toxicology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, 6525 HP Nijmegen, The Netherlands
| | - Carina Shayo
- the Laboratorio de Patología y Farmacología Molecular, Instituto de Biología y Medicina Experimental, 1428 Buenos Aires, Argentina
- the Consejo Nacional de Investigaciones Científicas y Técnicas and
| | - Carlos Davio
- From the Laboratorio de Farmacología de Receptores, Cátedra de Química Medicinal, Departamento de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, 1113 Buenos Aires, Argentina
- the Consejo Nacional de Investigaciones Científicas y Técnicas and
| |
Collapse
|
14
|
Côté SC, Pasvanis S, Bounou S, Dumais N. CCR7-specific migration to CCL19 and CCL21 is induced by PGE(2) stimulation in human monocytes: Involvement of EP(2)/EP(4) receptors activation. Mol Immunol 2009; 46:2682-93. [PMID: 19545899 DOI: 10.1016/j.molimm.2008.08.269] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2008] [Revised: 08/12/2008] [Accepted: 08/13/2008] [Indexed: 11/16/2022]
Abstract
The recent demonstration that newly recruited monocytes do not die at the site of inflammation, but migrate to draining lymph nodes, raises the question on the mechanism involved in this process. In this study, we demonstrate for the first time that prostaglandin E(2) (PGE(2)) regulates the expression and the activity of CCR7 in human blood-isolated monocytes as well as in the MONO-MAC-1 cell lineage. PGE(2) induces intracellular cAMP formation through engagement of the E-prostanoid 2/E-prostanoid 4 (EP(2)/EP(4)) receptors present on monocytes. Migration to chemokines CCL19 and CCL21 in the PGE(2)-stimulated monocytes is mediated through the augmentation of cAMP concentration and furthermore, the cAMP/PKA pathway appears to act as the major inducer of CCR7 transcription in MONO-MAC-1. While p38 MAPK was induced by PGE(2), we observed that PGE(2) can downregulate p42/p44 MAPK phosphorylation. At the transcription level, inhibition of p38 MAPK inhibits CCR7 mRNA expression. Finally, we demonstrated that transcription factors CREB-1 and C/EBPalpha and C/EBPbeta are translocated to the nucleus following PGE(2) stimulation and bind the potent CCR7 promoter region. Our findings may have important implication for HIV-1 migration to the lymph nodes since macrophages and monocytes, particularly CD16 positive subset, are susceptible to HIV-1 infection.
Collapse
Affiliation(s)
- Sandra C Côté
- Département de Biologie, Université de Sherbrooke, QC, Canada
| | | | | | | |
Collapse
|
15
|
Oevermann L, Scheitz J, Starke K, Köck K, Kiefer T, Dölken G, Niessen J, Greinacher A, Siegmund W, Zygmunt M, Kroemer HK, Jedlitschky G, Ritter CA. Hematopoietic stem cell differentiation affects expression and function of MRP4 (ABCC4), a transport protein for signaling molecules and drugs. Int J Cancer 2009; 124:2303-11. [PMID: 19142964 DOI: 10.1002/ijc.24207] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Several types of peripheral blood cells express ABC transporters. ABCC4 (MRP4) and ABCC5 (MRP5) localize to different cellular sites and fulfill lineage-specific functions such as mediator storage in platelets' dense granules. All mature blood cells originate from the same precursor and specific functionalities arise during differentiation. To characterize this process, expression, localization and function of MRP4 and MRP5 were assessed in differentiating human CD34+ progenitors and leukemia cell lines using real time polymerase chain reaction (PCR), immunofluorescence microscopy and cell viability assays. Median MRP4 mRNA copy numbers were significantly enhanced by megakaryocytic differentiation from 7.9 x 10(3) to 5.8 x 10(4) copies per nanograms of total RNA (p < 0.05) in CD34+ progenitors and in M-07e cells (MRP4 mRNA/18S rRNA ratios: 5.4 +/- 3.8 x 10(-4) vs. 2.7 +/- 0.9 x 10(-3) for native and differentiated cells, respectively, p < 0.05), and MRP4 protein was localized to granular structures and to the plasma membrane both in differentiated progenitors and bone marrow megakaryocytes. In contrast, expression of MRP4 decreased during maturation to leukocytes (MRP4 mRNA/18S rRNA ratios: 5.2 x 10(-3) for native vs. 3.5 x 10(-3) for CD34+ cells in the presence of G-CSF, p < 0.05) and was significantly reduced in mature monocytes and granulocytes compared with progenitors (MRP4 mRNA/18S rRNA ratios: 8.1 +/- 5.4 x 10(-5) and 2.8 +/- 1.6 x 10(-4) vs. 1.2 +/- 0.7 x 10(-3), respectively, p < 0.05). Expression of MRP5 was not significantly altered under all differentiation conditions. These results indicate that MRP4 expression is differentially regulated during hematopoiesis. The increase of MRP4 together with its specific localization during differentiation toward megakaryocytes supports the concept of platelet specific functions whereas decreased transporter expression in leukocyte differentiation may have implications for chemotherapy.
Collapse
Affiliation(s)
- Lena Oevermann
- Department of Pharmacology, Research Center of Pharmacology and Experimental Therapeutics, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Kim DC, Choi SY, Kim SH, Yun BS, Yoo ID, Reddy NRP, Yoon HS, Kim KT. Isoliquiritigenin selectively inhibits H(2) histamine receptor signaling. Mol Pharmacol 2006; 70:493-500. [PMID: 16675659 DOI: 10.1124/mol.106.023226] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Isoliquiritigenin, one of the major constituents of Glycyrrhiza uralensis (licorice), is a natural pigment with a simple chalcone structure 4,2',4'-trihydroxychalcone. In this study, isoliquiritigenin showed selective H(2) histamine receptor (H(2)R) antagonistic effect and remarkably reduced several H(2)R-mediated physiological responses. Preincubation of U937 and HL60 hematopoietic cells with isoliquiritigenin significantly inhibited H(2)R agonist-induced cAMP response in a concentration-dependent manner without affecting the viability of cells. Isoliquiritigenin also blocked the binding affinity of [(3)H]tiotidine to membrane receptors in HL-60 cells. Isoliquiritigenin did not affect the elevation of cAMP levels induced by cholera toxin, forskolin, or isoproterenol, indicating that the action site of isoliquiritigenin is not G(s) protein, effector enzyme, adenylyl cyclase, or beta(2)-adrenoceptor. Isoliquiritigenin affected neither H(1)R-nor H(3)R-mediated signaling. In molecular docking studies, isoliquiritigenin exhibited more favorable interactions with H(2)R than histamine. Isoliquiritigenin prominently inhibited H(2)R selective agonist dimaprit-induced cAMP generation in MKN-45 gastric cancer cell. Moreover, isoliquiritigenin reduced gastric acid secretion and protected gastric mucosal lesion formation in pylorus-ligated rat model. Taken together, the results demonstrate that isoliquiritigenin is an effective H(2)R antagonist and provides the basis for designing novel H(2)R antagonist.
Collapse
Affiliation(s)
- Dong-Chan Kim
- Department of Life Science, POSTECH, San 31, Hyoja Dong, Pohang 790-784, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Monczor F, Fernandez N, Riveiro E, Mladovan A, Baldi A, Shayo C, Davio C. Histamine H2 receptor overexpression induces U937 cell differentiation despite triggered mechanisms to attenuate cAMP signalling. Biochem Pharmacol 2006; 71:1219-28. [PMID: 16458858 DOI: 10.1016/j.bcp.2005.12.037] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2005] [Revised: 12/26/2005] [Accepted: 12/28/2005] [Indexed: 10/25/2022]
Abstract
Knowing that cell-surface receptors that recognize and respond to extracellular stimuli are key components for the regular communication between individual cells required for the survival of any living organism, the aim of the present work was to investigate the effect of H2R overexpression on the U937 signal transduction pathway and its consequences on cell proliferation and differentiation. The overexpression of H2R led to an increase in cAMP basal levels, a leftward shift of agonist concentration-response curves, and similar maximal response to agonist treatment, suggesting that overexpressed H2Rs act as functional spare receptors. In this system cells triggered several mechanisms tending to restore cAMP basal levels to those of the naïve cells. H2R overexpression induced PDE activity stimulation and GRK2 overexpression. In spite of the onset of these regulatory mechanisms, H2 agonist and rolipram treatments induced the terminal differentiation of the H2R overexpressed clone, conversely to the naïve cells. Present findings show that stably H2R overexpression alters cAMP signalling as the result of not only the amounts of second messenger generated but also the activation or upregulation of various components of signalling cascade, leading to an adapted biologically unique system. This adaptation may represent an advantage or a disadvantage, depending on the biological system, but in any case, the existence of compensatory mechanisms should be considered when a clinical treatment is designed.
Collapse
Affiliation(s)
- Federico Monczor
- Instituto de Biología y Medicina Experimental, Buenos Aires, Argentina.
| | | | | | | | | | | | | |
Collapse
|
18
|
Li L, Luo HS, Liu Y. Differences among desensitization of histamine H 2 receptor induced by three H 2 receptor antagonists on rat gastric parietal cells. Shijie Huaren Xiaohua Zazhi 2005; 13:2145-2147. [DOI: 10.11569/wcjd.v13.i17.2145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the differences of the desensitization of the histamine H2 receptor induced by three H2 receptor antagonists on the gastric parietal cells in rats.
METHODS: The gastric parietal cells were isolated by pronase digestion and then divided into three groups: cimetidine, ranitidine and famotidine treatment group. The activity of the H+-K+-ATPase was detected by H+-K+-ATPase kit after the cells were treated with different concentrations of the antagonists for different times.
RESULTS: Significant changes of the H+-K+-ATPase activity were observed after the cells were treated with different concentrations of the antagonists at different times. The activities of H+-K+-ATPase were significantly higher in famotidine group at 1, 2, and 4 h (589.34±2.7, 812.82±8.35, 637.15±4.59) than those in ranitidine (169.38±93.64, 343.46±44.88, 234.07±4.72) and cimetidine (118.42±5.91, 110.62±1.28, 102.43±3.44) group (P <0.01). The activity in ranitidine group was markedly higner than that in cimetidine group at 2 and 4 h. Famotidine increased the activities of H+-K+-ATPase significantly at the concentrations of 10 and 100 mg/L(178.21±20.38, 225.65±16.41) as compared with ranitidine and cimetidine did (70.88±21.44, 128.03±8.22; 123.62±4.32, 125.40±7.45)(P <0.01). At the concentration of 1000 mg/L, both famotidine and ranitidine increased the activity of H+-K+-ATPase obviously as compared with cimetidine did (233.44±6.24, 131.58±11.50 vs 109.88±0.69, P <0.01, P <0.05).
CONCLUSION: Three H2 receptor antagonists can induce different desensitization of the H2 receptor, among which famotidine induces the strongest and cimetidine does the weakest.
Collapse
|
19
|
Cohen M, Sklair-Tavron L, Pollack S. The Association of Forskolin-Stimulated Cyclic AMP Levels in Peripheral Blood Mononuclear Cells of Healthy People with Depressive Mood and Sense of Coherence. J PSYCHOPHYSIOL 2004. [DOI: 10.1027/0269-8803.18.4.177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Abstract: Background: Depressive mood exerts a suppressive effect on immune functions, mediated through neuro-immune processes. Research suggests a protective effect of sense of coherence (SOC) on psychological and physical health in face of stressful life events. Cyclic adenosine monophosphate (cAMP) is a cellular second messenger that is coupled to certain receptors expressed on peripheral blood mononuclear cells (PBMC). By interfering with the neuro-endocrine-immune axis, depressive mood may induce changes in intracellular cAMP levels in PBMC. Aims of study: To examine a possible association of SOC, stressful life events during the previous year, depressive mood, and cAMP levels. Methods: Fifteen male volunteers participated in the study. They completed the SOC and Zung's depression questionnaires and stressful life events inventory. Basic cAMP level and forskolin-stimulated cAMP production were measured in PBMC. Results: Forskolin-stimulated cAMP production was significantly higher in individuals who reported higher depressive mood, more stressful life events, or lower SOC scores. SOC scores explained 40% of forskolin-stimulated cAMP production. Basal cAMP levels could not be correlated with psychological scores. Age or education level did not affect forskolin-stimulated or basal cAMP levels. Conclusions: Our data suggest that high forskolin-stimulated cAMP production in PBMC is associated with low SOC, which points to availability of personal resources for coping with stressful life events. Further examination of the possible mediating role of SOC in relation to depressive mood and neuroimmune functions is needed.
Collapse
Affiliation(s)
- Miri Cohen
- Faculty of Social Welfare and Health Studies, Haifa University, Israel
- Department of Immunology, Rambam Medical Center and B. Rappaport Faculty of Medicine - Technion, Haifa, Israel
| | - Liora Sklair-Tavron
- Department of Immunology, Rambam Medical Center and B. Rappaport Faculty of Medicine - Technion, Haifa, Israel
- Innovative R&D, Teva Pharmaceutical Industries, Netanya, Israel
| | - Shimon Pollack
- Department of Immunology, Rambam Medical Center and B. Rappaport Faculty of Medicine - Technion, Haifa, Israel
| |
Collapse
|