1
|
Ribeiro IC, de Moraes JVB, Mariotini-Moura C, Polêto MD, da Rocha Torres Pavione N, de Castro RB, Miranda IL, Sartori SK, Alves KLS, Bressan GC, de Souza Vasconcellos R, Meyer-Fernandes JR, Diaz-Muñoz G, Fietto JLR. Synthesis of new non-natural L-glycosidic flavonoid derivatives and their evaluation as inhibitors of Trypanosoma cruzi ecto-nucleoside triphosphate diphosphohydrolase 1 (TcNTPDase1). Purinergic Signal 2024; 20:399-419. [PMID: 37975950 PMCID: PMC11303637 DOI: 10.1007/s11302-023-09974-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 10/20/2023] [Indexed: 11/19/2023] Open
Abstract
Trypanosoma cruzi is the pathogen of Chagas disease, a neglected tropical disease that affects more than 6 million people worldwide. There are no vaccines to prevent infection, and the therapeutic arsenal is very minimal and toxic. The unique E-NTPDase of T. cruzi (TcNTPDase1) plays essential roles in adhesion and infection and is a virulence factor. Quercetin is a flavonoid with antimicrobial, antiviral, and antitumor activities. Its potential as a partial inhibitor of NTPDases has also been demonstrated. In this work, we synthesized the non-natural L-glycoside derivatives of quercetin and evaluated them as inhibitors of recombinant TcNTPDase1 (rTcNTPDase1). These compounds, and quercetin and miquelianin, a natural quercetin derivative, were also tested. Compound 16 showed the most significant inhibitory effect (94%). Quercetin, miquelianin, and compound 14 showed inhibition close to 50%. We thoroughly investigated the inhibitory effect of 16. Our data suggested a competitive inhibition with a Ki of 8.39 μM (± 0.90). To better understand the interaction of compound 16 and rTcNTPDase1, we performed molecular dynamics simulations of the enzyme and docking analyses with the compounds. Our predictions show that compound 16 binds to the enzyme's catalytic site and interacts with important residues for NTPDase activity. As an inhibitor of a critical T. cruzi enzyme, (16) could be helpful as a starting point in the developing of a future treatment for Chagas disease. Furthermore, the discovery of (16) as an inhibitor of TcNTPDase1 may open new avenues in the study and development of new inhibitors of E-NTPDases.
Collapse
Affiliation(s)
- Isadora Cunha Ribeiro
- Biochemistry and Molecular Biology Department, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | | | - Christiane Mariotini-Moura
- General Biology Department, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
- Medicine and Nursing Department, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | - Marcelo Depolo Polêto
- Biochemistry and Molecular Biology Department, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | | | - Raissa Barbosa de Castro
- Biochemistry and Molecular Biology Department, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | - Izabel Luzia Miranda
- Exact Science Institute, Chemistry Department, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Suélen Karine Sartori
- Exact Science Institute, Chemistry Department, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Kryssia Lohayne Santos Alves
- Exact Science Institute, Chemistry Department, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Gustavo Costa Bressan
- Biochemistry and Molecular Biology Department, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | | | - José Roberto Meyer-Fernandes
- Laboratory of Cellular Biochemistry, Institute of Medical Biochemistry Leopoldo de Meis, Health Sciences Center, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gaspar Diaz-Muñoz
- Exact Science Institute, Chemistry Department, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| | - Juliana Lopes Rangel Fietto
- Biochemistry and Molecular Biology Department, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil.
- General Biology Department, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil.
| |
Collapse
|
2
|
Spatola BN, Lerner AG, Wong C, Dela Cruz T, Welch M, Fung W, Kovalenko M, Losenkova K, Yegutkin GG, Beers C, Corbin J, Soros VB. Fully human anti-CD39 antibody potently inhibits ATPase activity in cancer cells via uncompetitive allosteric mechanism. MAbs 2021; 12:1838036. [PMID: 33146056 PMCID: PMC7646477 DOI: 10.1080/19420862.2020.1838036] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The extracellular ATP/adenosine axis in the tumor microenvironment (TME) has emerged as an important immune-regulatory pathway. Nucleoside triphosphate diphosphohydrolase-1 (NTPDase1), otherwise known as CD39, is highly expressed in the TME, both on infiltrating immune cells and tumor cells across a broad set of cancer indications. CD39 processes pro-inflammatory extracellular ATP to ADP and AMP, which is then processed by Ecto-5ʹ-nucleotidase/CD73 to immunosuppressive adenosine. Directly inhibiting the enzymatic function of CD39 via an antibody has the potential to unleash an immune-mediated anti-tumor response via two mechanisms: 1) increasing the availability of immunostimulatory extracellular ATP released by damaged and/or dying cells, and 2) reducing the generation and accumulation of suppressive adenosine within the TME. Tizona Therapeutics has engineered a novel first-in-class fully human anti-CD39 antibody, TTX-030, that directly inhibits CD39 ATPase enzymatic function with sub-nanomolar potency. Further characterization of the mechanism of inhibition by TTX-030 using CD39+ human melanoma cell line SK-MEL-28 revealed an uncompetitive allosteric mechanism (α < 1). The uncompetitive mechanism of action enables TTX-030 to inhibit CD39 at the elevated ATP concentrations reported in the TME. Maximal inhibition of cellular CD39 ATPase velocity was 85%, which compares favorably to results reported for antibody inhibitors to other enzyme targets. The allosteric mechanism of TTX-030 was confirmed via mapping the epitope to a region of CD39 distant from its active site, which suggests possible models for how potent inhibition is achieved. In summary, TTX-030 is a potent allosteric inhibitor of CD39 ATPase activity that is currently being evaluated in clinical trials for cancer therapy.
Collapse
Affiliation(s)
- Bradley N Spatola
- Antibody Development, Tizona Therapeutics , South San Francisco, CA, USA
| | - Alana G Lerner
- Immunology, Tizona Therapeutics , South San Francisco, CA, USA
| | - Clifford Wong
- Antibody Development, Tizona Therapeutics , South San Francisco, CA, USA
| | - Tracy Dela Cruz
- Immunology, Tizona Therapeutics , South San Francisco, CA, USA.,Immunology, Trishula Therapeutics, South San Francisco , CA, USA
| | - Megan Welch
- Immunology, Tizona Therapeutics , South San Francisco, CA, USA
| | - Wanchi Fung
- Antibody Development, Tizona Therapeutics , South San Francisco, CA, USA
| | | | | | | | - Courtney Beers
- Immunology, Tizona Therapeutics , South San Francisco, CA, USA
| | - John Corbin
- Antibody Development, Tizona Therapeutics , South San Francisco, CA, USA
| | - Vanessa B Soros
- Antibody Development, Tizona Therapeutics , South San Francisco, CA, USA
| |
Collapse
|
3
|
Zeng J, Ning Z, Wang Y, Xiong H. Implications of CD39 in immune-related diseases. Int Immunopharmacol 2020; 89:107055. [PMID: 33045579 DOI: 10.1016/j.intimp.2020.107055] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/10/2020] [Accepted: 09/26/2020] [Indexed: 02/07/2023]
Abstract
Extracellular adenosine triphosphate (eATP) mediates pro-inflammatory responses by recruiting and activating inflammatory cells. CD39 can hydrolyze eATP into adenosine monophosphate (AMP), while CD73 can convert AMP into the immunosuppressive nucleoside adenosine (ADO). CD39 is a rate-limiting enzyme in this cascade, which is regarded as an immunological switch shifting the ATP-mediated pro-inflammatory environment to the ADO- mediated anti-inflammatory status. The CD39 expression can be detected in a wide spectrum of immunocytes, which is under the influence of environmental and genetic factors. It is increasingly suggested that, CD39 participates in some pathophysiological processes, like inflammatory bowel disease (IBD), sepsis, multiple sclerosis (MS), allergic diseases, ischemia-reperfusion (I/R) injury, systemic lupus erythematosus (SLE), diabetes and cancer. Here, we focus on the current understanding of CD39 in immunity, and comprehensively illustrate the diverse CD39 functions within a variety of disorders.
Collapse
Affiliation(s)
- Jianrui Zeng
- Institute of Immunology and Molecular Medicine, Jining Medical University, Shandong 272067, China
| | - Zhaochen Ning
- Institute of Immunology and Molecular Medicine, Jining Medical University, Shandong 272067, China
| | - Yuzhong Wang
- Department of Neurology and Central Laboratory, Affiliated Hospital of Jining Medical University, Shandong 272000, China.
| | - Huabao Xiong
- Institute of Immunology and Molecular Medicine, Jining Medical University, Shandong 272067, China.
| |
Collapse
|
4
|
Krieger AC, Anand R, Hernandez-Rosa E, Maidman A, Milrad S, DeGrazia MQ, Choi AJ, Oromendia C, Marcus AJ, Drosopoulos JHF. Increased platelet activation in sleep apnea subjects with intermittent hypoxemia. Sleep Breath 2020; 24:1537-1547. [PMID: 32036486 DOI: 10.1007/s11325-020-02021-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 01/10/2020] [Accepted: 01/27/2020] [Indexed: 01/30/2023]
Abstract
PURPOSE Obstructive sleep apnea (OSA) is independently associated with increased risk for stroke and other cardiovascular diseases. Since activated platelets play an important role in cardiovascular disease, the objective of this study was to determine whether platelet reactivity was altered in OSA subjects with intermittent nocturnal hypoxemia. METHODS Thirty-one subjects, without hypertension or cardiovascular disease and not taking medication, participated in the study. Subjects were stratified based on OSA-related oxygen desaturation index (ODI) recorded during overnight polysomnography. Platelet reactivity to a broad panel of agonists (collagen, thrombin, protease-activated receptor1 hexapeptide, epinephrine, ADP) was measured by monitoring platelet aggregation and ATP secretion. Expression of platelet activation markers CD154 (CD40L) and CD62P (P-selectin) and platelet-monocyte aggregates (PMA) was quantified by flow cytometry. RESULTS Epinephrine-induced platelet aggregation was substantially decreased in OSA subjects with significant intermittent hypoxemia (ODI ≥ 15) compared with subjects with milder hypoxemia levels (ODI < 15) (area under curve, p = 0.01). In addition, OSA subjects with ODI ≥ 15 exhibited decreased thrombin-induced platelet aggregation (p = 0.02) and CD40L platelet surface expression (p = 0.05). Platelet responses to the other agonists, CD62P platelet surface expression, and PMA levels were not significantly different between groups. Reduction in platelet responses to epinephrine and thrombin, and decreased CD40L surface marker expression in significant hypoxemic OSA individuals, is consistent with their platelets being in an activated state. CONCLUSIONS Increased platelet activation was present in otherwise healthy subjects with intermittent nocturnal hypoxemia due to underlying OSA. This prothrombotic milieu in the vasculature is likely a key contributing factor toward development of thrombosis and cardiovascular disease. TRIAL REGISTRATION NCT00859950.
Collapse
Affiliation(s)
- Ana C Krieger
- Department of Neurology, Weill Cornell Medicine, New York, NY, 10065, USA.,Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Ranjini Anand
- Thrombosis Research Laboratory, Research Service, VA New York Harbor Healthcare System, 423 East 23rd Street, Room 13026W, New York, NY, 10010, USA.,Department of Medicine, Division of Hematology/Oncology, Weill Cornell Medicine, New York, NY, 10065, USA.,Medical Department, PureSinse Inc., Mississauga, ON, L4W 5K4, Canada
| | - Evelyn Hernandez-Rosa
- Thrombosis Research Laboratory, Research Service, VA New York Harbor Healthcare System, 423 East 23rd Street, Room 13026W, New York, NY, 10010, USA.,Department of Medicine, Division of Hematology/Oncology, Weill Cornell Medicine, New York, NY, 10065, USA.,Department of Pathology, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, 10032, USA
| | - Allison Maidman
- Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA.,Department of Pediatrics, NYU Langone Hospital, Brooklyn, NY, 11220, USA
| | - Sara Milrad
- Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA.,Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, 33431, USA
| | - Miles Q DeGrazia
- Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Alexander J Choi
- Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA.,University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Clara Oromendia
- Department of Healthcare Policy and Research, Division of Biostatistics and Epidemiology, Weill Cornell Medicine, New York, NY, 10065, USA.,Project Rōnin Inc., San Mateo, CA, 94401, USA
| | - Aaron J Marcus
- Thrombosis Research Laboratory, Research Service, VA New York Harbor Healthcare System, 423 East 23rd Street, Room 13026W, New York, NY, 10010, USA.,Department of Medicine, Division of Hematology/Oncology, Weill Cornell Medicine, New York, NY, 10065, USA.,Department of Pathology and Laboratory Medicine, Division of Hematology/Oncology, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Joan H F Drosopoulos
- Thrombosis Research Laboratory, Research Service, VA New York Harbor Healthcare System, 423 East 23rd Street, Room 13026W, New York, NY, 10010, USA. .,Department of Medicine, Division of Hematology/Oncology, Weill Cornell Medicine, New York, NY, 10065, USA.
| |
Collapse
|
5
|
Characterization of soluble CD39 (SolCD39/NTPDase1) from PiggyBac nonviral system as a tool to control the nucleotides level. Biochem J 2019; 476:1637-1651. [PMID: 31085558 DOI: 10.1042/bcj20190040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 05/10/2019] [Accepted: 05/13/2019] [Indexed: 11/17/2022]
Abstract
Extracellular ATP (eATP) and its metabolites have emerged as key modulators of different diseases and comprise a complex pathway called purinergic signaling. An increased number of tools have been developed to study the role of nucleotides and nucleosides in cell proliferation and migration, influence on the immune system and tumor progression. These tools include receptor agonists/antagonists, engineered ectonucleotidases, interference RNAs and ectonucleotidase inhibitors that allow the control and quantification of nucleotide levels. NTPDase1 (also called apyrase, ecto-ATPase and CD39) is one of the main enzymes responsible for the hydrolysis of eATP, and purified enzymes, such as apyrase purified from potato, or engineered as soluble CD39 (SolCD39), have been widely used in in vitro and in vivo experiments. However, the commercial apyrase had its effects recently questioned and SolCD39 exhibits limitations, such as short half-life and need of high doses to reach the expected enzymatic activity. Therefore, this study investigated a non-viral method to improve the overexpression of SolCD39 and evaluated its impact on other enzymes of the purinergic system. Our data demonstrated that PiggyBac transposon system proved to be a fast and efficient method to generate cells stably expressing SolCD39, producing high amounts of the enzyme from a limited number of cells and with high hydrolytic activity. In addition, the soluble form of NTPDase1/CD39 did not alter the expression or catalytic activity of other enzymes from the purinergic system. Altogether, these findings set the groundwork for prospective studies on the function and therapeutic role of eATP and its metabolites in physiological and pathological conditions.
Collapse
|
6
|
Lee SY, Luo X, Namasivayam V, Geiss J, Mirza S, Pelletier J, Stephan H, Sévigny J, Müller CE. Development of a selective and highly sensitive fluorescence assay for nucleoside triphosphate diphosphohydrolase1 (NTPDase1, CD39). Analyst 2019; 143:5417-5430. [PMID: 30303204 DOI: 10.1039/c8an01108g] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Ecto-nucleoside triphosphate diphosphohydrolase1 (NTPDase1, CD39) is a major ectonucleotidase that hydrolyzes proinflammatory ATP via ADP to AMP, which is subsequently converted by ecto-5'-nucleotidase (CD73) to immunosuppressive adenosine. Activation of CD39 has potential for treating inflammatory diseases, while inhibition was suggested as a novel strategy for the immunotherapy of cancer. In the present study, we developed a selective and highly sensitive capillary electrophoresis (CE) assay using a novel fluorescent CD39 substrate, a fluorescein-labelled ATP (PSB-170621A) that is converted to its AMP derivative. To accelerate the assays, a two-directional (forward and reverse) CE system was implemented using 96-well plates, which is suitable for the screening of compound libraries (Z'-factor: 0.7). The detection limits for the forward and reverse operation were 11.7 and 2.00 pM, respectively, indicating a large enhancement in sensitivity as compared to previous methods (e.g. malachite-green assay: 1 000 000-fold, CE-UV assay: 500 000-fold, fluorescence polarization immunoassay: 12 500-fold). Enzyme kinetic studies at human CD39 revealed a Km value of 19.6 μM, and a kcat value of 119 × 10-3 s-1 for PSB-170621A, which shows similar substrate properties as ATP (11.4 μM and 82.5 × 10-3 s-1). The compound displayed similar properties at rat and mouse CD39. Subsequent docking studies into a homology model of human CD39 revealed a hydrophobic pocket that accommodates the fluorescein tag. PSB-170621A was found to be preferably hydrolyzed by CD39 as compared to other ectonucleotidases. The new assay was validated by performing inhibition assays with several standard CD39 inhibitors yielding results that were consonant with data using the natural substrates.
Collapse
Affiliation(s)
- Sang-Yong Lee
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
7
|
|
8
|
Molecular dynamic simulations reveal structural insights into substrate and inhibitor binding modes and functionality of Ecto-Nucleoside Triphosphate Diphosphohydrolases. Sci Rep 2018; 8:2581. [PMID: 29416085 PMCID: PMC5803232 DOI: 10.1038/s41598-018-20971-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 01/26/2018] [Indexed: 11/25/2022] Open
Abstract
Ecto-nucleotidase enzymes catalyze the hydrolysis of extracellular nucleotides to their respective nucleosides. Herein, we place the focus on the elucidation of structural features of the cell surface located ecto-nucleoside triphosphate diphosphohydrolases (E-NTPDase1-3 and 8). The physiological role of these isozymes is crucially important as they control purinergic signaling by modulating the extracellular availability of nucleotides. Since, crystal or NMR structure of the human isozymes are not available – structures have been obtained by homology modeling. Refinement of the homology models with poor stereo-chemical quality is of utmost importance in order to derive reliable structures for subsequent studies. Therefore, the resultant models obtained by homology modelling were refined by running molecular dynamic simulation. Binding mode analysis of standard substrates and of competitive inhibitor was conducted to highlight important regions of the active site involved in hydrolysis of the substrates and possible mechanism of inhibition.
Collapse
|
9
|
Fuentes E, Palomo I. Extracellular ATP metabolism on vascular endothelial cells: A pathway with pro-thrombotic and anti-thrombotic molecules. Vascul Pharmacol 2015; 75:1-6. [PMID: 25989108 DOI: 10.1016/j.vph.2015.05.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2015] [Revised: 04/16/2015] [Accepted: 05/09/2015] [Indexed: 12/20/2022]
Abstract
Vascular endothelial contributes to the metabolism and interconversion of extracellular adenine nucleotides via ecto-ATPase/ADPase (CD39) and ecto-5'nucleotidase (CD73) activities. These enzymes collectively dephosphorylate ATP, ADP, and AMP with the production of additional adenosine. In the vascular system, adenine nucleotides (ATP and ADP) and nucleoside adenosine represent an important class of extracellular molecules involved in modulating the processes linked to vascular thrombosis exerting various effects in platelets. Yet, the mechanisms by which the extracellular ATP metabolism in the local environment trigger pro-thrombotic and anti-thrombotic states are yet to be fully elucidated. In this article, the relative contribution of extracellular ATP metabolism in platelet regulation is explored.
Collapse
Affiliation(s)
- Eduardo Fuentes
- Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule, R09I2001 Talca, Chile.
| | - Iván Palomo
- Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile; Centro de Estudios en Alimentos Procesados (CEAP), CONICYT-Regional, Gore Maule, R09I2001 Talca, Chile.
| |
Collapse
|
10
|
Fujii S, Fujimoto K, Goto N, Kanawa M, Kawamoto T, Pan H, Srivatanakul P, Rakdang W, Pornprasitwech J, Saskianti T, Suardita K, Nishimura F, Kato Y. Characteristic expression of MSX1, MSX2, TBX2 and ENTPD1 in dental pulp cells. Biomed Rep 2015; 3:566-572. [PMID: 26171167 DOI: 10.3892/br.2015.456] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 03/20/2015] [Indexed: 01/08/2023] Open
Abstract
Dental pulp cells (DPCs) are a promising source of transplantable cells in regenerative medicine. However, DPCs have not been fully characterized at the molecular level. The aim of the present study was to distinguish DPCs from various source-derived mesenchymal stem cells (MSCs), fibroblasts (FBs) and other cells by the expression of several DPC-characteristic genes. DPCs were isolated from human pulp tissues by the explant method or the enzyme digestion method, and maintained with media containing 10% serum or 7.5% platelet-rich plasma. RNA was isolated from the cells and from dental pulp tissue specimens. The mRNA levels were determined by DNA microarray and quantitative polymerase chain reaction analyses. The msh homeobox 1, msh homeobox 2, T-box 2 and ectonucleoside triphosphate diphosphohydrolase 1 mRNA levels in DPCs were higher than that of the levels identified in the following cell types: MSCs derived from bone marrow, synovium and adipose tissue; and in cells such as FBs, osteoblasts, adipocytes and chondrocytes. The enhanced expression in DPCs was consistently observed irrespective of donor age, tooth type and culture medium. In addition, these genes were expressed at high levels in dental pulp tissue in vivo. In conclusion, this gene set may be useful in the identification and characterization of DPCs in basic studies and pulp cell-based regeneration therapy.
Collapse
Affiliation(s)
- Sakiko Fujii
- Department of Dental Science for Health Promotion, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima 734-8553, Japan
| | - Katsumi Fujimoto
- Department of Dental and Medical Biochemistry, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima 734-8553, Japan
| | - Noriko Goto
- Department of Pediatric Dentistry, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima 734-8553, Japan
| | - Masami Kanawa
- Natural Science Center for Basic Research and Development, Hiroshima University, Hiroshima, Hiroshima 734-8553, Japan
| | - Takeshi Kawamoto
- Department of Dental and Medical Biochemistry, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima 734-8553, Japan
| | - Haiou Pan
- Two Cells Co., Ltd., Hiroshima, Hiroshima 734-8551, Japan
| | | | - Waralak Rakdang
- BioEden Asia Co., Ltd., Klong Luang, Pathum Thani 12120, Thailand
| | | | - Tania Saskianti
- Department of Pediatric Dentistry, Airlangga University, Surabaya, East Java 60132, Indonesia
| | - Ketut Suardita
- Department of Conservative Dentistry, Airlangga University, Surabaya, East Java 60132, Indonesia
| | - Fusanori Nishimura
- Department of Dental Science for Health Promotion, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima 734-8553, Japan
| | - Yukio Kato
- Department of Dental and Medical Biochemistry, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima 734-8553, Japan
| |
Collapse
|
11
|
Moeckel D, Jeong SS, Sun X, Broekman MJ, Nguyen A, Drosopoulos JHF, Marcus AJ, Robson SC, Chen R, Abendschein D. Optimizing human apyrase to treat arterial thrombosis and limit reperfusion injury without increasing bleeding risk. Sci Transl Med 2015; 6:248ra105. [PMID: 25100739 DOI: 10.1126/scitranslmed.3009246] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In patients with acute myocardial infarction undergoing reperfusion therapy to restore blood flow through blocked arteries, simultaneous inhibition of platelet P2Y12 receptors with the current standard of care neither completely prevents recurrent thrombosis nor provides satisfactory protection against reperfusion injury. Additionally, these antiplatelet drugs increase the risk of bleeding. To devise a different strategy, we engineered and optimized the apyrase activity of human nucleoside triphosphate diphosphohydrolase-3 (CD39L3) to enhance scavenging of extracellular adenosine diphosphate, a predominant ligand of P2Y12 receptors. The resulting recombinant protein, APT102, exhibited greater than four times higher adenosine diphosphatase activity and a 50 times longer plasma half-life than did native apyrase. Treatment with APT102 before coronary fibrinolysis with intravenous recombinant human tissue-type plasminogen activator in conscious dogs completely prevented thrombotic reocclusion and significantly decreased infarction size by 81% without increasing bleeding time. In contrast, clopidogrel did not prevent coronary reocclusion and increased bleeding time. In a murine model of myocardial reperfusion injury caused by transient coronary artery occlusion, APT102 also decreased infarct size by 51%, whereas clopidogrel was not effective. These preclinical data suggest that APT102 should be tested for its ability to safely and effectively maximize the benefits of myocardial reperfusion therapy in patients with arterial thrombosis.
Collapse
Affiliation(s)
- Douglas Moeckel
- Center for Cardiovascular Research, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Soon Soeg Jeong
- APT Therapeutics Inc., 4041 Forest Park Avenue, St. Louis, MO 63108, USA
| | - Xiaofeng Sun
- Division of Gastroenterology/Hepatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - M Johan Broekman
- Thrombosis Research Laboratory, Research Service, Veterans Affairs New York Harbor Healthcare System, New York, NY 10010, USA. Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Annie Nguyen
- Center for Cardiovascular Research, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joan H F Drosopoulos
- Thrombosis Research Laboratory, Research Service, Veterans Affairs New York Harbor Healthcare System, New York, NY 10010, USA. Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Aaron J Marcus
- Thrombosis Research Laboratory, Research Service, Veterans Affairs New York Harbor Healthcare System, New York, NY 10010, USA. Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Simon C Robson
- Division of Gastroenterology/Hepatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Ridong Chen
- APT Therapeutics Inc., 4041 Forest Park Avenue, St. Louis, MO 63108, USA.
| | - Dana Abendschein
- Center for Cardiovascular Research, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
12
|
Nunes VS, Vasconcelos EG, Faria-Pinto P, Borges CCH, Capriles PVSZ. Structural Comparative Analysis of Ecto- NTPDase Models from S. Mansoni and H. Sapiens. ACTA ACUST UNITED AC 2015. [DOI: 10.1007/978-3-319-19048-8_21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
|
13
|
Okuhata R, Otsuka Y, Tsuchiya T, Kanzawa N. Mutagenesis of apyrase conserved region 1 alters the nucleotide substrate specificity. PLANT SIGNALING & BEHAVIOR 2013; 8:e24131. [PMID: 23470725 PMCID: PMC3908943 DOI: 10.4161/psb.24131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 02/27/2013] [Indexed: 05/29/2023]
Abstract
Two apyrases having different substrate specificity, MP67 and MpAPY2, are present in Mimosa pudica. The substrate specificity of MP67 is quite high against ADP, and is distinct from any other apyrase. This might be attributed to the nucleotide binding motif (DXG) in apyrase conserved region 1. We performed a single amino acid substitution at position X in the motif. The ratio of the velocity of ATP/ADP hydrolysis was higher (approximately 1) for the S63A-MP67 mutant than for wild type-MP67 (0.19). Binding affinity for ADP of A75S-MpAPY2 mutant was increased to a level higher than that of the wild type MpAPY2. Thus, the residue at position X in the DXG motif plays an important role in determining nucleotide preference.
Collapse
|
14
|
Leite PM, Gomes RS, Figueiredo AB, Serafim TD, Tafuri WL, de Souza CC, Moura SAL, Fietto JLR, Melo MN, Ribeiro-Dias F, Oliveira MAP, Rabello A, Afonso LCC. Ecto-nucleotidase activities of promastigotes from Leishmania (Viannia) braziliensis relates to parasite infectivity and disease clinical outcome. PLoS Negl Trop Dis 2012; 6:e1850. [PMID: 23071853 PMCID: PMC3469556 DOI: 10.1371/journal.pntd.0001850] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Accepted: 08/22/2012] [Indexed: 12/22/2022] Open
Abstract
Background Leishmania (Viannia) braziliensis has been associated with a broad range of clinical manifestations ranging from a simple cutaneous ulcer to destructive mucosal lesions. Factors leading to this diversity of clinical presentations are not clear, but parasite factors have lately been recognized as important in determining disease progression. Given the fact that the activity of ecto-nucleotidases correlates with parasitism and the development of infection, we evaluated the activity of these enzymes in promastigotes from 23 L. braziliensis isolates as a possible parasite-related factor that could influence the clinical outcome of the disease. Methodology/Principal Findings Our results show that the isolates differ in their ability to hydrolyze adenine nucleotides. Furthermore, we observed a positive correlation between the time for peak of lesion development in C57BL/6J mice and enzymatic activity and clinical manifestation of the isolate. In addition, we found that L. (V.) braziliensis isolates obtained from mucosal lesions hydrolyze higher amounts of adenine nucleotides than isolates obtained from skin lesions. One isolate with high (PPS6m) and another with low (SSF) ecto-nucleotidase activity were chosen for further studies. Mice inoculated with PPS6m show delayed lesion development and present larger parasite loads than animals inoculated with the SSF isolate. In addition, PPS6m modulates the host immune response by inhibiting dendritic cell activation and NO production by activated J774 macrophages. Finally, we observed that the amastigote forms from PPS6m and SSF isolates present low enzymatic activity that does not interfere with NO production and parasite survival in macrophages. Conclusions/Significance Our data suggest that ecto-nucleotidases present on the promastigote forms of the parasite may interfere with the establishment of the immune response with consequent impaired ability to control parasite dissemination and this may be an important factor in determining the clinical outcome of leishmaniasis. Cutaneous leishmaniasis is a widespread tropical disease caused by different species of Leishmania protozoa that are transmitted by infected sandflies. Clinical presentations are extremely diverse and dependent on a variety of parasite and host factors that are poorly understood. Leishmania (V.) braziliensis infection may result in a devastating disease manifestation characterized by the development of destructive lesions in the oral, nasal, and pharyngeal mucosal. Ecto-nucleotidases are enzymes that are involved in the hydrolysis of extracellular nucleotides. These enzymes have been shown to correlate with virulence of Leishmania parasites. In this work, we evaluated the ecto-nucleotidase activity of promastigotes from the twenty three different L. braziliensis isolates. We demonstrated that isolates obtained from mucosal lesions present higher levels of ecto-nucleotidase activity than those from cutaneous lesions. In addition, we show that in the murine model of cutaneous leishmaniasis, promastigote forms of parasite with higher activity induce a delayed/decreased immune response that may correlate with spreading of the parasites throughout the body. Thus, we propose that the level of ecto-nucleotidase activity of promastigotes may be a marker for the development of severe clinical forms of cutaneous leishmaniasis and also a possible target for future therapeutic intervention.
Collapse
Affiliation(s)
- Pauline M. Leite
- Laboratório de Imunoparasitologia, DECBI/NUPEB, Universidade Federal de Ouro Preto, Minas Gerais, Brazil
| | - Rodrigo S. Gomes
- Laboratório de Imunoparasitologia, DECBI/NUPEB, Universidade Federal de Ouro Preto, Minas Gerais, Brazil
| | - Amanda B. Figueiredo
- Laboratório de Imunoparasitologia, DECBI/NUPEB, Universidade Federal de Ouro Preto, Minas Gerais, Brazil
| | - Tiago D. Serafim
- Laboratório de Imunoparasitologia, DECBI/NUPEB, Universidade Federal de Ouro Preto, Minas Gerais, Brazil
| | - Wagner L. Tafuri
- Departamento de Patologia Geral, ICB, UFMG, Belo Horizonte, Minas Gerais, Brazil
| | - Carolina C. de Souza
- Departamento de Patologia Geral, ICB, UFMG, Belo Horizonte, Minas Gerais, Brazil
| | - Sandra A. L. Moura
- Laboratório de Imunoparasitologia, DECBI/NUPEB, Universidade Federal de Ouro Preto, Minas Gerais, Brazil
| | - Juliana L. R. Fietto
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Viçosa, Minas Gerais, Brazil
| | - Maria N. Melo
- Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fátima Ribeiro-Dias
- Instituto de Patologia Tropical e Saúde Publica, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Milton A. P. Oliveira
- Instituto de Patologia Tropical e Saúde Publica, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Ana Rabello
- Centro de Pesquisas René Rachou- FIOCRUZ, Belo Horizonte, Minas Gerais, Brazil
| | - Luís C. C. Afonso
- Laboratório de Imunoparasitologia, DECBI/NUPEB, Universidade Federal de Ouro Preto, Minas Gerais, Brazil
- * E-mail:
| |
Collapse
|
15
|
Zimmermann H, Zebisch M, Sträter N. Cellular function and molecular structure of ecto-nucleotidases. Purinergic Signal 2012; 8:437-502. [PMID: 22555564 PMCID: PMC3360096 DOI: 10.1007/s11302-012-9309-4] [Citation(s) in RCA: 786] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Accepted: 02/01/2012] [Indexed: 12/12/2022] Open
Abstract
Ecto-nucleotidases play a pivotal role in purinergic signal transmission. They hydrolyze extracellular nucleotides and thus can control their availability at purinergic P2 receptors. They generate extracellular nucleosides for cellular reuptake and salvage via nucleoside transporters of the plasma membrane. The extracellular adenosine formed acts as an agonist of purinergic P1 receptors. They also can produce and hydrolyze extracellular inorganic pyrophosphate that is of major relevance in the control of bone mineralization. This review discusses and compares four major groups of ecto-nucleotidases: the ecto-nucleoside triphosphate diphosphohydrolases, ecto-5'-nucleotidase, ecto-nucleotide pyrophosphatase/phosphodiesterases, and alkaline phosphatases. Only recently and based on crystal structures, detailed information regarding the spatial structures and catalytic mechanisms has become available for members of these four ecto-nucleotidase families. This permits detailed predictions of their catalytic mechanisms and a comparison between the individual enzyme groups. The review focuses on the principal biochemical, cell biological, catalytic, and structural properties of the enzymes and provides brief reference to tissue distribution, and physiological and pathophysiological functions.
Collapse
Affiliation(s)
- Herbert Zimmermann
- Institute of Cell Biology and Neuroscience, Molecular and Cellular Neurobiology, Biologicum, Goethe-University Frankfurt, Max-von-Laue-Str. 13, 60438, Frankfurt am Main, Germany.
| | | | | |
Collapse
|
16
|
Krug U, Zebisch M, Krauss M, Sträter N. Structural insight into activation mechanism of Toxoplasma gondii nucleoside triphosphate diphosphohydrolases by disulfide reduction. J Biol Chem 2011; 287:3051-66. [PMID: 22130673 DOI: 10.1074/jbc.m111.294348] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The intracellular parasite Toxoplasma gondii produces two nucleoside triphosphate diphosphohydrolases (NTPDase1 and -3). These tetrameric, cysteine-rich enzymes require activation by reductive cleavage of a hitherto unknown disulfide bond. Despite a 97% sequence identity, both isozymes differ largely in their ability to hydrolyze ATP and ADP. Here, we present crystal structures of inactive NTPDase3 as an apo form and in complex with the product AMP to resolutions of 2.0 and 2.2 Å, respectively. We find that the enzyme is present in an open conformation that precludes productive substrate binding and catalysis. The cysteine bridge 258-268 is identified to be responsible for locking of activity. Crystal structures of constitutively active variants of NTPDase1 and -3 generated by mutation of Cys(258)-Cys(268) show that opening of the regulatory cysteine bridge induces a pronounced contraction of the whole tetramer. This is accompanied by a 12° domain closure motion resulting in the correct arrangement of all active site residues. A complex structure of activated NTPDase3 with a non-hydrolyzable ATP analog and the cofactor Mg(2+) to a resolution of 2.85 Å indicates that catalytic differences between the NTPDases are primarily dictated by differences in positioning of the adenine base caused by substitution of Arg(492) and Glu(493) in NTPDase1 by glycines in NTPDase3.
Collapse
Affiliation(s)
- Ulrike Krug
- Institute of Bioanalytical Chemistry, Center for Biotechnology and Biomedicine, University of Leipzig, D-04103 Leipzig, Germany
| | | | | | | |
Collapse
|
17
|
Thammavongsa V, Schneewind O, Missiakas DM. Enzymatic properties of Staphylococcus aureus adenosine synthase (AdsA). BMC BIOCHEMISTRY 2011; 12:56. [PMID: 22035583 PMCID: PMC3213008 DOI: 10.1186/1471-2091-12-56] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Accepted: 10/28/2011] [Indexed: 01/08/2023]
Abstract
Background Staphylococcus aureus is a human pathogen that produces extracellular adenosine to evade clearance by the host immune system, an activity attributed to the 5'-nucleotidase activity of adenosine synthase (AdsA). In mammals, conversion of adenosine triphosphate to adenosine is catalyzed in a two-step process: ecto-nucleoside triphosphate diphosphohydrolases (ecto-NTDPases) hydrolyze ATP and ADP to AMP, whereas 5'-nucleotidases hydrolyze AMP to adenosine. NTPDases harbor apyrase conserved regions (ACRs) that are critical for activity. Results NTPDase ACR motifs are absent in AdsA, yet we report here that recombinant AdsA hydrolyzes ADP and ATP in addition to AMP. Competition assays suggest that hydrolysis occurs following binding of all three substrates at a unique site. Alanine substitution of two amino acids, aspartic acid 127 and histidine 196 within the 5'-nucleotidase signature sequence, leads to reduced AMP or ADP hydrolysis but does not affect the binding of these substrates. Conclusion Collectively, these results provide insight into the unique ability of AdsA to produce adenosine through the consecutive hydrolysis of ATP, ADP and AMP, thereby endowing S. aureus with the ability to modulate host immune responses.
Collapse
Affiliation(s)
- Vilasack Thammavongsa
- Department of Microbiology, University of Chicago, 920 E, 58th St, Chicago, Illinois 60637, USA
| | | | | |
Collapse
|
18
|
Kirley TL, Crawford PA, Smith TM. The structure of the nucleoside triphosphate diphosphohydrolases (NTPDases) as revealed by mutagenic and computational modeling analyses. Purinergic Signal 2011; 2:379-89. [PMID: 17710224 PMCID: PMC1950141 DOI: 10.1007/s11302-005-5301-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Over the last seven years our laboratory has focused on the determination of the structural aspects of nucleoside triphosphate diphosphohydrolases (NTPDases) using site-directed mutagenesis and computational comparative protein modeling to generate hypotheses and models for the hydrolytic site and enzymatic mechanism of the family of NTPDase nucleotidases. This review summarizes these studies utilizing NTPDase3 (also known as CD39L3 and HB6), an NTPDase family member that is intermediate in its characteristics between the more widely distributed and studied NTPDase1 (also known as CD39) and NTPDase2 (also known as CD39L1 and ecto-ATPase) enzymes. Relevant site-directed mutagenesis studies of other NTPDases are also discussed and compared to NTPDase3 results. It is anticipated that many of the results and conclusions reached via studies of NTPDase3 will be relevant to understanding the structure and enzymatic mechanism of all the cell-surface members of this family (NTPDase1–3, 8), and that understanding these NTPDase enzymes will aid in modulating the many varied processes under purinergic signaling control. This review also integrates the site-directed mutagenesis results with a recent 3-D structural model for the extracellular portion of NTPDases that helps explain the importance of the apyrase conserved regions (ACRs) of the NTPDases. Utilizing this model and published work from Dr Guidotti’s laboratory concerning the importance and characteristics of the two transmembrane helices and their movements in response to substrate, we present a speculative cartoon model of the enzymatic mechanism of the membrane-bound NTPDases that integrates movements of the extracellular region required for catalysis with movements of the N- and C-terminal transmembrane helices that are important for control and modulation of enzyme activity.
Collapse
Affiliation(s)
- Terence L. Kirley
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, 231 Albert Sabin Way, Cincinnati, Ohio 45267-0575 USA
| | - Patrick A. Crawford
- Department of Pharmacology and Cell Biophysics, University of Cincinnati, College of Medicine, 231 Albert Sabin Way, Cincinnati, Ohio 45267-0575 USA
| | - Thomas M. Smith
- Department of Cardiovascular and Metabolic Diseases, Wyeth Research, Cambridge, Massachusetts USA
| |
Collapse
|
19
|
Papanikolaou A, Papafotika A, Christoforidis S. CD39 Reveals Novel Insights into the Role of Transmembrane Domains in Protein Processing, Apical Targeting and Activity. Traffic 2011; 12:1148-65. [DOI: 10.1111/j.1600-0854.2011.01224.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
20
|
Tanaka K, Nguyen CT, Libault M, Cheng J, Stacey G. Enzymatic activity of the soybean ecto-apyrase GS52 is essential for stimulation of nodulation. PLANT PHYSIOLOGY 2011; 155:1988-98. [PMID: 21346172 PMCID: PMC3091080 DOI: 10.1104/pp.110.170910] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2010] [Accepted: 02/19/2011] [Indexed: 05/20/2023]
Abstract
Nitrogen is an essential nutrient for plant growth. In the Rhizobium-legume symbiosis, root nodules are the sites of bacterial nitrogen fixation, in which atmospheric nitrogen is converted into a form that plants can utilize. While recent studies suggested an important role for the soybean (Glycine max) ecto-apyrase GS52 in rhizobial root hair infection and root nodule formation, precisely how this protein impacts the nodulation process remains undetermined. In this study, the biochemical characteristics of the GS52 enzyme were investigated. Computer modeling of the GS52 apyrase structure identified key amino acid residues important for catalytic activity, which were subsequently mutagenized. Although the GS52 enzyme exhibited broad substrate specificity, its activity on pyrimidine nucleotides and diphosphate nucleotides was significantly higher than on ATP. This result was corroborated by structural modeling of GS52, which predicted a low specificity for the adenine base within the substrate-binding pocket of the enzyme. The wild-type enzyme and its inactive mutant forms were expressed in soybean roots in order to evaluate the importance of GS52 enzymatic activity for nodulation. The results indicated a clear correlation between GS52 enzymatic activity and nodule number. Altogether, our study indicates that the catalytic activity of the GS52 apyrase, likely acting on extracellular nucleotides, is critical for rhizobial infection and nodulation.
Collapse
Affiliation(s)
| | | | | | | | - Gary Stacey
- Division of Plant Sciences, C.S. Bond Life Sciences Center (K.T., C.T.N., M.L., G.S.), National Center for Soybean Biotechnology (K.T., M.L., G.S.), Department of Computer Science, Informatics Institute (J.C.), and Division of Biochemistry (G.S.), University of Missouri, Columbia, Missouri 65211
| |
Collapse
|
21
|
Knowles AF. The GDA1_CD39 superfamily: NTPDases with diverse functions. Purinergic Signal 2011; 7:21-45. [PMID: 21484095 DOI: 10.1007/s11302-010-9214-7] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2010] [Accepted: 12/21/2010] [Indexed: 01/05/2023] Open
Abstract
The first comprehensive review of the ubiquitous "ecto-ATPases" by Plesner was published in 1995. A year later, a lymphoid cell activation antigen, CD39, that had been cloned previously, was shown to be an ecto-ATPase. A family of proteins, related to CD39 and a yeast GDPase, all containing the canonical apyrase conserved regions in their polypeptides, soon started to expand. They are now recognized as members of the GDA1_CD39 protein family. Because proteins in this family hydrolyze nucleoside triphosphates and diphosphates, a unifying nomenclature, nucleoside triphosphate diphopshohydrolases (NTPDases), was established in 2000. Membrane-bound NTPDases are either located on the cell surface or membranes of intracellular organelles. Soluble NTPDases exist in the cytosol and may be secreted. In the last 15 years, molecular cloning and functional expression have facilitated biochemical characterization of NTPDases of many organisms, culminating in the recent structural determination of the ecto-domain of a mammalian cell surface NTPDase and a bacterial NTPDase. The first goal of this review is to summarize the biochemical, mutagenesis, and structural studies of the NTPDases. Because of their ability in hydrolyzing extracellular nucleotides, the mammalian cell surface NTPDases (the ecto-NTPDases) which regulate purinergic signaling have received the most attention. Less appreciated are the functions of intracellular NTPDases and NTPDases of other organisms, e.g., bacteria, parasites, Drosophila, plants, etc. The second goal of this review is to summarize recent findings which demonstrate the involvement of the NTPDases in multiple and diverse physiological processes: pathogen-host interaction, plant growth, eukaryote cell protein and lipid glycosylation, eye development, and oncogenesis.
Collapse
Affiliation(s)
- Aileen F Knowles
- Department of Chemistry and Biochemistry, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182-1030, USA,
| |
Collapse
|
22
|
Vivian JP, Riedmaier P, Ge H, Le Nours J, Sansom FM, Wilce MCJ, Byres E, Dias M, Schmidberger JW, Cowan PJ, d'Apice AJF, Hartland EL, Rossjohn J, Beddoe T. Crystal structure of a Legionella pneumophila ecto -triphosphate diphosphohydrolase, a structural and functional homolog of the eukaryotic NTPDases. Structure 2010; 18:228-38. [PMID: 20159467 DOI: 10.1016/j.str.2009.11.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2009] [Revised: 11/16/2009] [Accepted: 11/24/2009] [Indexed: 12/20/2022]
Abstract
Many pathogenic bacteria have sophisticated mechanisms to interfere with the mammalian immune response. These include the disruption of host extracellular ATP levels that, in humans, is tightly regulated by the nucleoside triphosphate diphosphohydrolase family (NTPDases). NTPDases are found almost exclusively in eukaryotes, the notable exception being their presence in some pathogenic prokaryotes. To address the function of bacterial NTPDases, we describe the structures of an NTPDase from the pathogen Legionella pneumophila (Lpg1905/Lp1NTPDase) in its apo state and in complex with the ATP analog AMPPNP and the subtype-specific NTPDase inhibitor ARL 67156. Lp1NTPDase is structurally and catalytically related to eukaryotic NTPDases and the structure provides a basis for NTPDase-specific inhibition. Furthermore, we demonstrate that the activity of Lp1NTPDase correlates directly with intracellular replication of Legionella within macrophages. Collectively, these findings provide insight into the mechanism of this enzyme and highlight its role in host-pathogen interactions.
Collapse
Affiliation(s)
- Julian P Vivian
- The Protein Crystallography Unit, ARC Centre of Excellence in Structural and Functional Microbial Genomics, Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, Victoria 3800, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Gaddie KJ, Kirley TL. Proline residues link the active site to transmembrane domain movements in human nucleoside triphosphate diphosphohydrolase 3 (NTPDase3). Purinergic Signal 2010; 6:327-37. [PMID: 21103216 DOI: 10.1007/s11302-010-9180-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2009] [Accepted: 03/01/2010] [Indexed: 10/19/2022] Open
Abstract
The active sites of the membrane-bound nucleoside triphosphate diphosphohydrolases (NTPDases) regulate and are regulated by coordinated and spatially distant movements of their transmembrane helices, modulating enzyme activity, and substrate specificity. Using site-directed mutagenesis, the roles of the conserved proline residues (N-terminal: P52 and P53; C-terminal: P472, P476, P481, P484, and P485) of human NTPDase3, located in the "linker regions" that connect the N- and C-terminal transmembrane helices with the extracellular active site, were examined. Single cysteine substitutions were strategically placed in the transmembrane domain (N-terminal helix: V42C; C-terminal helix: G489C) to serve as cross-linking "sensors" of helical interactions. These "sensor" background mutant proteins (V42C and G489C NTPDase3) are enzymatically active and are cross-linked by copper phenanthroline less efficiently in the presence of adenosine triphosphate (ATP). Proline to alanine substitutions at P53, P481, P484, and P485 in the V42C background, as well as P53, P481, and P484 in the G489C background, exhibited decreased nucleotidase activities. More importantly, alanine substitutions at P53 and P481 in the V42C background and P481 in the G489C background no longer exhibited the ATP-induced decrease in transmembrane cross-linking efficiency. Interestingly, the P485A mutation abolished oxidative cross-linking at G489C both in the presence and absence of ATP. Taken together, these results suggest a role for proline residues 53 and 481 in the linker regions of human NTPDase3 for coupling nucleotide binding at the enzyme active site to movements and/or rearrangements of the transmembrane helices necessary for optimal nucleotide hydrolysis.
Collapse
Affiliation(s)
- Keith J Gaddie
- Department of Pharmacology and Cell Biophysics, College of Medicine, University of Cincinnati, P.O. Box 670575, Cincinnati, OH 45267-0575 USA
| | | |
Collapse
|
24
|
Li CS, Lee Y, Knowles AF. The stability of chicken nucleoside triphosphate diphosphohydrolase 8 requires both of its transmembrane domains. Biochemistry 2010; 49:134-46. [PMID: 20000380 DOI: 10.1021/bi901820c] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Chicken nucleoside triphosphate diphosphohydrolase 8 (NTPDase8) is a cell surface ectonucleotidase with a large extracellular domain (ECD) containing the active site and is anchored to the membrane by two transmembrane domains (TMDs) at the N- and C-termini. Unlike other cell surface NTPDases that have been characterized, the chicken NTPDase8 is not susceptible to substrate inactivation or agents that cause membrane perturbation. To determine if the stability of the enzyme is inherent in its ECD, the cDNA construct of the soluble chicken NTPDase8 was expressed and the protein purified. The ATPase activity of the purified soluble chicken NTPDase8 was less than 15% of that of the purified full-length enzyme. Strikingly, in contrast to the membrane-bound enzyme, the activity of the soluble chicken NTPDase8 decreased with time in a temperature-dependent manner as a result of inactivation by ATP, ADP, and P(i). Truncated mutants in which the ECD is anchored by a single TMD at either the N- or the C-terminus by the native chicken NTPDase TMDs or a TMD from a different NTPDase, human NTPDase2, also displayed a nonlinear time course of ATP hydrolysis. While removal of the N- or C-terminal TMD affected protein expression differently, the truncated mutants were generally similar to the soluble chicken NTPDase8 with respect to ATP, ADP, and P(i) inactivation. Other biochemical characteristics, e.g., ATPase/ADPase ratios, inhibition by azide, and affinity for ATP, were also altered when one or both of the TMDs were removed from the chicken NTPDase8. These results indicate that (1) both TMDs of the chicken NTPDase8 are required to maintain stability of the enzyme and maximal catalytic activity and (2) the conformations of the ectodomain in the soluble enzyme and the truncated mutants differ from that of the full-length chicken NTPDase8.
Collapse
Affiliation(s)
- Cheryl S Li
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, California 92182-1030, USA
| | | | | |
Collapse
|
25
|
Drosopoulos JHF, Kraemer R, Shen H, Upmacis RK, Marcus AJ, Musi E. Human solCD39 inhibits injury-induced development of neointimal hyperplasia. Thromb Haemost 2009; 103:426-34. [PMID: 20024507 DOI: 10.1160/th09-05-0305] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2009] [Accepted: 11/01/2009] [Indexed: 12/13/2022]
Abstract
Blood platelets provide the initial response to vascular endothelial injury, becoming activated as they adhere to the injured site. Activated platelets recruit leukocytes, and initiate proliferation and migration of vascular smooth muscle cells (SMC) within the injured vessel wall, leading to development of neointimal hyperplasia. Endothelial CD39/NTPDase1 and recombinant solCD39 rapidly metabolise nucleotides, including stimulatory ADP released from activated platelets, thereby suppressing additional platelet reactivity. Using a murine model of vascular endothelial injury, we investigated whether circulating human solCD39 could reduce platelet activation and accumulation, thus abating leukocyte infiltration and neointimal formation following vascular damage. Intraperitoneally-administered solCD39 ADPase activity in plasma peaked 1 hour (h) post-injection, with an elimination half-life of 43 h. Accordingly, mice were administered solCD39 or saline 1 h prior to vessel injury, then either sacrificed 24 h post-injury or treated with solCD39 or saline (three times weekly) for an additional 18 days. Twenty-four hours post-injury, solCD39-treated mice displayed a reduction in platelet activation and recruitment, P-selectin expression, and leukocyte accumulation in the arterial lumen. Furthermore, repeated administration of solCD39 modulated the late stage of vascular injury by suppressing leukocyte deposition, macrophage infiltration and smooth muscle cell (SMC) proliferation/migration, resulting in abrogation of neointimal thickening. In contrast, injured femoral arteries of saline-injected mice exhibited massive platelet thrombus formation, marked P-selectin expression, and leukocyte infiltration. Pronounced neointimal growth with macrophage and SMC accretion was also observed (intimal-to-medial area ratio 1.56 +/- 0.34 at 19 days). Thus, systemic administration of solCD39 profoundly affects injury-induced cellular responses, minimising platelet deposition and leukocyte recruitment, and suppressing neointimal hyperplasia.
Collapse
Affiliation(s)
- J H F Drosopoulos
- Thrombosis Research Laboratory, Room 13026W, VA New York Harbor Healthcare System, 423 East 23rd Street, New York, N.Y. 10010-5050, USA.
| | | | | | | | | | | |
Collapse
|
26
|
Possible effects of microbial ecto-nucleoside triphosphate diphosphohydrolases on host-pathogen interactions. Microbiol Mol Biol Rev 2009; 72:765-81, Table of Contents. [PMID: 19052327 DOI: 10.1128/mmbr.00013-08] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In humans, purinergic signaling plays an important role in the modulation of immune responses through specific receptors that recognize nucleoside tri- and diphosphates as signaling molecules. Ecto-nucleoside triphosphate diphosphohydrolases (ecto-NTPDases) have important roles in the regulation of purinergic signaling by controlling levels of extracellular nucleotides. This process is key to pathophysiological protective responses such as hemostasis and inflammation. Ecto-NTPDases are found in all higher eukaryotes, and recently it has become apparent that a number of important parasitic pathogens of humans express surface-located NTPDases that have been linked to virulence. For those parasites that are purine auxotrophs, these enzymes may play an important role in purine scavenging, although they may also influence the host response to infection. Although ecto-NTPDases are rare in bacteria, expression of a secreted NTPDase in Legionella pneumophila was recently described. This ecto-enzyme enhances intracellular growth of the bacterium and potentially affects virulence. This discovery represents an important advance in the understanding of the contribution of other microbial NTPDases to host-pathogen interactions. Here we review other progress made to date in the characterization of ecto-NTPDases from microbial pathogens, how they differ from mammalian enzymes, and their association with organism viability and virulence. In addition, we postulate how ecto-NTPDases may contribute to the host-pathogen interaction by reviewing the effect of selected microbial pathogens on purinergic signaling. Finally, we raise the possibility of targeting ecto-NTPDases in the development of novel anti-infective agents based on potential structural and clear enzymatic differences from the mammalian ecto-NTPDases.
Collapse
|
27
|
Structural insight into signal conversion and inactivation by NTPDase2 in purinergic signaling. Proc Natl Acad Sci U S A 2008; 105:6882-7. [PMID: 18458329 DOI: 10.1073/pnas.0802535105] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cell surface-located nucleoside triphosphate diphosphohydrolases (NTPDase1, -2, -3, and -8) are oligomeric integral membrane proteins responsible for signal conversion and inactivation in extracellular nucleotide-mediated "purinergic" signaling. They catalyze the sequential hydrolysis of the signaling molecule ATP via ADP to AMP. Here we present the structure of the extracellular domain of Rattus norvegicus NTPDase2 in an active state at resolutions between 1.7 A and 2.1 A in four different forms: (i) apo form, (ii) ternary complex with the nonhydrolyzable ATP analog AMPPNP and cofactor Ca(2+), (iii) quaternary complex with Ca(2+) and bound products AMP and phosphate, and (iv) binary product complex with AMP only. Analysis of the ATP (analog) binding mode explains the importance of several residues for activity and allows suggestion of a catalytic mechanism. The carboxylate group of E165 serves as a catalytic base and activates a water molecule, which is well positioned for nucleophilic attack on the terminal phosphate. Based on analysis of the two product complex structures in which AMP adopts different conformations, a substrate binding mode for ADP hydrolysis is proposed. This allows for an understanding of how the same hydrolytic site can be engaged in ATP and ADP but not AMP hydrolysis.
Collapse
|
28
|
Sansom FM, Riedmaier P, Newton HJ, Dunstone MA, Müller CE, Stephan H, Byres E, Beddoe T, Rossjohn J, Cowan PJ, d'Apice AJF, Robson SC, Hartland EL. Enzymatic properties of an ecto-nucleoside triphosphate diphosphohydrolase from Legionella pneumophila: substrate specificity and requirement for virulence. J Biol Chem 2008; 283:12909-18. [PMID: 18337253 DOI: 10.1074/jbc.m801006200] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Legionella pneumophila is the predominant cause of Legionnaires disease, a severe and potentially fatal form of pneumonia. Recently, we identified an ecto-nucleoside triphosphate diphosphohydrolase (NTPDase) from L. pneumophila, termed Lpg1905, which enhances intracellular replication of L. pneumophila in eukaryotic cells. Lpg1905 is the first prokaryotic member of the CD39/NTPDase1 family of enzymes, which are characterized by the presence of five apyrase conserved regions and the ability to hydrolyze nucleoside tri- and diphosphates. Here we examined the substrate specificity of Lpg1905 and showed that apart from ATP and ADP, the enzyme catalyzed the hydrolysis of GTP and GDP but had limited activity against CTP, CDP, UTP, and UDP. Based on amino acid residues conserved in the apyrase conserved regions of eukaryotic NTPDases, we generated five site-directed mutants, Lpg1905E159A, R122A, N168A, Q193A, and W384A. Although the mutations E159A, R122A, Q193A, and W384A abrogated activity completely, N168A resulted in decreased activity caused by reduced affinity for nucleotides. When introduced into the lpg1905 mutant strain of L. pneumophila, only N168A partially restored the ability of L. pneumophila to replicate in THP-1 macrophages. Following intratracheal inoculation of A/J mice, none of the Lpg1905 mutants was able to restore virulence to an lpg1905 mutant during lung infection, thereby demonstrating the importance of NTPDase activity to L. pneumophila infection. Overall, the kinetic studies undertaken here demonstrated important differences to mammalian NTPDases and different sensitivities to NTPDase inhibitors that may reflect underlying structural variations.
Collapse
Affiliation(s)
- Fiona M Sansom
- Department of Microbiology and Immunology and Medicine, University of Melbourne, Parkville, Victoria 3010, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Uccelletti D, Pascoli A, Farina F, Alberti A, Mancini P, Hirschberg CB, Palleschi C. APY-1, a novel Caenorhabditis elegans apyrase involved in unfolded protein response signalling and stress responses. Mol Biol Cell 2008; 19:1337-45. [PMID: 18216284 DOI: 10.1091/mbc.e07-06-0547] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Protein glycosylation modulates a wide variety of intracellular events and dysfunction of the glycosylation pathway has been reported in a variety of human pathologies. Endo-apyrases have been suggested to have critical roles in protein glycosylation and sugar metabolism. However, deciphering the physiological relevance of Endo-apyrases activity has actually proved difficult, owing to their complexity and the functional redundancy within the family. We report here that a UDP/GDPase, homologous to the human apyrase Scan-1, is present in the membranes of Caenorhabditis elegans, encoded by the ORF F08C6.6 and hereinafter-named APY-1. We showed that ER stress induced by tunicamycin or high temperature resulted in increased transcription of apy-1. This increase was not observed in C. elegans mutants defective in ire-1 or atf-6, demonstrating the requirement of both ER stress sensors for up-regulation of apy-1. Depletion of APY-1 resulted in constitutively activated unfolded protein response. Defects in the pharynx and impaired organization of thin fibers in muscle cells were observed in adult worms depleted of APY-1. Some of the apy-1(RNAi) phenotypes are suggestive of premature aging, because these animals also showed accumulation of lipofuscin and reduced lifespan that was not dependent on the functioning of DAF-2, the receptor of the insulin/IGF-1 signaling pathway.
Collapse
Affiliation(s)
- D Uccelletti
- Department of Developmental and Cell Biology, University of Rome "La Sapienza," 00185 Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
30
|
Pulte ED, Broekman MJ, Olson KE, Drosopoulos JHF, Kizer JR, Islam N, Marcus AJ. CD39/NTPDase-1 activity and expression in normal leukocytes. Thromb Res 2007; 121:309-17. [PMID: 17555802 PMCID: PMC2255570 DOI: 10.1016/j.thromres.2007.04.008] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2007] [Revised: 04/17/2007] [Accepted: 04/24/2007] [Indexed: 10/23/2022]
Abstract
INTRODUCTION CD39/NTPDase-1 is a cell surface enzyme expressed on leukocytes and endothelial cells that metabolizes ATP to ADP and AMP. CD39 is expressed on numerous different types of normal leukocytes, but details of its expression have not been determined previously. METHODS We examined CD39 expression and activity in leukocytes isolated from healthy volunteers. Expression of CD39 on leukocytes was measured by FACS and activity of CD39 in lymphocytes and neutrophils was determined by an enzymatic radio-TLC assay. RESULTS We established that CD39 is expressed on neutrophils, lymphocytes, and monocytes. The enzyme is found on >90% of monocytes, neutrophils, and B-lymphocytes, and 6% of T-lymphocytes and natural killer cells. Per cell density of expression varied, with the highest expression on monocytes and B-lymphocytes. ATPase and ADPase activities were highest on B-lymphocytes, lower on neutrophils, lowest on T-lymphocytes. The ratio of ADPase:ATPase activity was 1.8 for neutrophils and B-lymphocytes and 1.4 for T-lymphocytes. Hypertensive volunteers had lower levels of CD39 on their T-lymphocytes and NK cells. No correlation between age, gender, ethnic background, or cholesterol level and CD39 expression was observed. CONCLUSIONS We conclude that CD39 activity and expression are present to varying degrees on all leukocytes types examined. Differences between leukocyte types should be considered when examining CD39 in disease states.
Collapse
Affiliation(s)
- E Dianne Pulte
- Research Service, Veterans Affairs New York Harbor Healthcare System, New York, NY 10010
- Medicine-Hematology/Oncology, Weill Medical College of Cornell University, New York, NY 10021
| | - M Johan Broekman
- Research Service, Veterans Affairs New York Harbor Healthcare System, New York, NY 10010
- Medicine-Hematology/Oncology, Weill Medical College of Cornell University, New York, NY 10021
| | - Kim E Olson
- Medicine-Hematology/Oncology, Weill Medical College of Cornell University, New York, NY 10021
| | - Joan H F Drosopoulos
- Research Service, Veterans Affairs New York Harbor Healthcare System, New York, NY 10010
- Medicine-Hematology/Oncology, Weill Medical College of Cornell University, New York, NY 10021
| | - Jorge R Kizer
- Medicine and Public Health, Weill Medical College of Cornell University, New York, NY 10021
| | - Naziba Islam
- Research Service, Veterans Affairs New York Harbor Healthcare System, New York, NY 10010
- Medicine-Hematology/Oncology, Weill Medical College of Cornell University, New York, NY 10021
| | - Aaron J Marcus
- Research Service, Veterans Affairs New York Harbor Healthcare System, New York, NY 10010
- Medicine-Hematology/Oncology, Weill Medical College of Cornell University, New York, NY 10021
- Pathology and Laboratory Medicine, Weill Medical College of Cornell University, New York, NY 10021
| |
Collapse
|
31
|
Pulte D, Olson KE, Broekman MJ, Islam N, Ballard HS, Furman RR, Olson AE, Marcus AJ. CD39 activity correlates with stage and inhibits platelet reactivity in chronic lymphocytic leukemia. J Transl Med 2007; 5:23. [PMID: 17480228 PMCID: PMC1885243 DOI: 10.1186/1479-5876-5-23] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2007] [Accepted: 05/04/2007] [Indexed: 01/10/2023] Open
Abstract
Background Chronic lymphocytic leukemia (CLL) is characterized by accumulation of mature appearing lymphocytes and is rarely complicated by thrombosis. One possible explanation for the paucity of thrombotic events in these patients may be the presence of the ecto-nucleotidase CD39/NTDPase-1 on the surface of the malignant cells in CLL. CD39 is the major promoter of platelet inhibition in vivo via its metabolism of ADP to AMP. We hypothesize that if CD39 is observed on CLL cells, then patients with CLL may be relatively protected against platelet aggregation and recruitment and that CD39 may have other effects on CLL, including modulation of the disease, via its metabolism of ATP. Methods Normal and malignant lymphocytes were isolated from whole blood from patients with CLL and healthy volunteers. Enzyme activity was measured via radio-TLC assay and expression via FACS. Semi-quantititative RT-PCR for CD39 splice variants and platelet function tests were performed on several samples. Results Functional assays demonstrated that ADPase and ATPase activities were much higher in CLL cells than in total lymphocytes from the normal population on a per cell basis (p-value < 0.00001). CD39 activity was elevated in stage 0–2 CLL compared to stage 3–4 (p < 0.01). FACS of lymphocytes demonstrated CD39 expression on > 90% of normal and malignant B-lymphocytes and ~8% of normal T-lymphocytes. RT-PCR showed increased full length CD39 and splice variant 1.5, but decreased variant 1.3 in CLL cells. Platelet function tests showed inhibition of platelet activation and recruitment to ADP by CLL cells. Conclusion CD39 is expressed and active on CLL cells. Enzyme activity is higher in earlier stages of CLL and decreased enzyme activity may be associated with worsening disease. These results suggest that CD39 may play a role in the pathogenesis of malignancy and protect CLL patients from thrombotic events.
Collapse
MESH Headings
- Adenosine Diphosphate/pharmacology
- Adult
- Aged
- Aged, 80 and over
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Apyrase/genetics
- Apyrase/metabolism
- B-Lymphocytes/metabolism
- B-Lymphocytes/pathology
- Chromatography, Thin Layer
- Female
- Flow Cytometry
- Gene Expression Regulation, Leukemic/drug effects
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/physiopathology
- Lymphocyte Subsets/metabolism
- Male
- Middle Aged
- Platelet Activation/drug effects
- Platelet Activation/physiology
- Platelet Aggregation/drug effects
- Reverse Transcriptase Polymerase Chain Reaction
- T-Lymphocytes/metabolism
- T-Lymphocytes/pathology
Collapse
Affiliation(s)
- Dianne Pulte
- Research Service, Veterans Affairs New York Harbor Healthcare System, New York, NY 10010, USA
- Medicine-Hematology/Oncology, Weill Medical College Cornell University, New York, NY 10021, USA
| | - Kim E Olson
- Research Service, Veterans Affairs New York Harbor Healthcare System, New York, NY 10010, USA
- Medicine-Hematology/Oncology, Weill Medical College Cornell University, New York, NY 10021, USA
| | - M Johan Broekman
- Research Service, Veterans Affairs New York Harbor Healthcare System, New York, NY 10010, USA
- Medicine-Hematology/Oncology, Weill Medical College Cornell University, New York, NY 10021, USA
| | - Naziba Islam
- Research Service, Veterans Affairs New York Harbor Healthcare System, New York, NY 10010, USA
- Medicine-Hematology/Oncology, Weill Medical College Cornell University, New York, NY 10021, USA
| | - Harold S Ballard
- Medical Service, VA NY Harbor Healtcare System, New York, NY 10010, USA
| | - Richard R Furman
- Medicine-Hematology/Oncology, Weill Medical College Cornell University, New York, NY 10021, USA
| | - Ashley E Olson
- Research Service, Veterans Affairs New York Harbor Healthcare System, New York, NY 10010, USA
| | - Aaron J Marcus
- Research Service, Veterans Affairs New York Harbor Healthcare System, New York, NY 10010, USA
- Medicine-Hematology/Oncology, Weill Medical College Cornell University, New York, NY 10021, USA
- Medical Service, VA NY Harbor Healtcare System, New York, NY 10010, USA
- Pathology and Laboratory Medicine, Weill Medical College of Cornell University, New York, NY 10021, USA
| |
Collapse
|
32
|
Sansom FM, Newton HJ, Crikis S, Cianciotto NP, Cowan PJ, d'Apice AJF, Hartland EL. A bacterial ecto-triphosphate diphosphohydrolase similar to human CD39 is essential for intracellular multiplication of Legionella pneumophila. Cell Microbiol 2007; 9:1922-35. [PMID: 17388784 DOI: 10.1111/j.1462-5822.2007.00924.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
As part of its pathogenesis, Legionella pneumophila persists within human alveolar macrophages in non-acidified organelles that do not mature into phagolysosomes. Two L. pneumophila genes, lpg0971 and lpg1905, are predicted to encode ecto-nucleoside triphosphate diphosphohydrolases (ecto-NTPDases) that share sequence similarity with human CD39/NTPDase1. The predicted products possess five apyrase conserved domains that are typical of eukaryotic ecto-NTPDases. In this study, we found that an lpg1905 mutant was recovered in lower numbers from macrophages, alveolar epithelial cells and the amoeba, Hartmannella vermiformis compared with wild-type L. pneumophila and an lpg0971 mutant. Similar to human CD39, recombinant purified Lpg1905 exhibited ATPase and ADPase activity and possessed the ability to inhibit platelet aggregation. Mutation of a conserved Glu159 residue that is essential for CD39 activity inhibited ATPase and ADPase activity of Lpg1905. In addition, enzyme activity was inhibited in the presence of the specific ecto-NTPDase inhibitor, ARL67156. The entry and replication defect of the lpg1905 mutant was reversed upon transcomplementation with lpg1905 but not lpg1905E159A encoding an enzymatically inactive form of the protein. Although several protozoan parasites exhibit ecto-NTPDase activity, including Toxoplasma gondii, Trichomonas vaginalis and Trypanosoma cruzi, this is the first time a bacterial ecto-NTPDase has been implicated in virulence.
Collapse
Affiliation(s)
- Fiona M Sansom
- Australian Bacterial Pathogenesis Program, Department of Microbiology, Monash University, Victoria 3800, Australia
| | | | | | | | | | | | | |
Collapse
|
33
|
Grinthal A, Guidotti G. Bilayer mechanical properties regulate the transmembrane helix mobility and enzymatic state of CD39. Biochemistry 2007; 46:279-90. [PMID: 17198399 PMCID: PMC2536646 DOI: 10.1021/bi061052p] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
CD39 can exist in at least two distinct functional states depending on the presence and intact membrane integration of its two transmembrane helices. In native membranes, the transmembrane helices undergo dynamic rotational motions that are required for enzymatic activity and are regulated by substrate binding. In this study, we show that bilayer mechanical properties regulate conversion between the two enzymatic functional states by modulating transmembrane helix dynamics. Alteration of membrane properties by insertion of cone-shaped or inverse cone-shaped amphiphiles or by cholesterol removal switches CD39 to the same enzymatic state that removal or solubilization of the transmembrane domains does. The same membrane alterations increase the propensity of both transmembrane helices to rotate within the packed structure, resulting in a structure with greater mobility but not an altered primary conformation. Membrane alteration also abolishes the ability of the substrate to stabilize the helices in their primary conformation, indicating a loss of coupling between substrate binding and transmembrane helix dynamics. Removal of either transmembrane helix mimics the effect of membrane alteration on the mobility and substrate sensitivity of the remaining helix, suggesting that the ends of the extracellular domain have intrinsic flexibility. We suggest that a mechanical bilayer property, potentially elasticity, regulates CD39 by altering the balance between the stability and flexibility of its transmembrane helices and, in turn, of its active site.
Collapse
Affiliation(s)
- Alison Grinthal
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts 02138, USA.
| | | |
Collapse
|
34
|
Musi E, Islam N, Drosopoulos JHF. Constraints imposed by transmembrane domains affect enzymatic activity of membrane-associated human CD39/NTPDase1 mutants. Arch Biochem Biophys 2007; 461:30-9. [PMID: 17374358 DOI: 10.1016/j.abb.2007.02.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2006] [Revised: 02/02/2007] [Accepted: 02/03/2007] [Indexed: 01/30/2023]
Abstract
Human CD39/NTPDase1 is an endothelial cell membrane-associated nucleotidase. Its large extracellular domain rapidly metabolizes nucleotides, especially ADP released from activated platelets, inhibiting further platelet activation/recruitment. Previous studies using our recombinant soluble CD39 demonstrated the importance of residues S57, D54, and D213 for enzymatic/biological activity. We now report effects of S57A, D54A, and D213A mutations on full-length (FL)CD39 function. Enzymatic activity of alanine modified FLCD39s was less than wild-type, contrasting the enhanced activity of their soluble counterparts. Furthermore, conservative substitutions D54E and D213E led to enzymes with activities greater than the alanine modified FLCD39s, but less than wild-type. Reductions in mutant activities were primarily associated with reduced catalytic rates. Differences in enzymatic activity were not attributable to gross changes in the nucleotide binding pocket or the enzyme's ability to multimerize. Thus, composition of the active site of wild-type CD39 appears optimized for ADPase function in the context of the transmembrane domains.
Collapse
Affiliation(s)
- Elgilda Musi
- Thrombosis Research Laboratory, Research Service, Room 13026W, VA New York Harbor Healthcare System, 423 East 23rd Street, New York, NY 10010-5050, USA
| | | | | |
Collapse
|
35
|
Kopp HG, Hooper AT, Broekman MJ, Avecilla ST, Petit I, Luo M, Milde T, Ramos CA, Zhang F, Kopp T, Bornstein P, Jin DK, Marcus AJ, Rafii S. Thrombospondins deployed by thrombopoietic cells determine angiogenic switch and extent of revascularization. J Clin Invest 2007; 116:3277-91. [PMID: 17143334 PMCID: PMC1679710 DOI: 10.1172/jci29314] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2006] [Accepted: 10/24/2006] [Indexed: 11/17/2022] Open
Abstract
Thrombopoietic cells may differentially promote or inhibit tissue vascularization by releasing both pro- and antiangiogenic factors. However, the molecular determinants controlling the angiogenic phenotype of thrombopoietic cells remain unknown. Here, we show that expression and release of thrombospondins (TSPs) by megakaryocytes and platelets function as a major antiangiogenic switch. TSPs inhibited thrombopoiesis, diminished bone marrow microvascular reconstruction following myelosuppression, and limited the extent of revascularization in a model of hind limb ischemia. We demonstrate that thrombopoietic recovery following myelosuppression was significantly enhanced in mice deficient in both TSP1 and TSP2 (TSP-DKO mice) in comparison with WT mice. Megakaryocyte and platelet levels in TSP-DKO mice were rapidly restored, thereby accelerating revascularization of myelosuppressed bone marrow and ischemic hind limbs. In addition, thrombopoietic cells derived from TSP-DKO mice were more effective in supporting neoangiogenesis in Matrigel plugs. The proangiogenic activity of TSP-DKO thrombopoietic cells was mediated through activation of MMP-9 and enhanced release of stromal cell-derived factor 1. Thus, TSP-deficient thrombopoietic cells function as proangiogenic agents, accelerating hemangiogenesis within the marrow and revascularization of ischemic hind limbs. As such, interference with the release of cellular stores of TSPs may be clinically effective in augmenting neoangiogenesis.
Collapse
Affiliation(s)
- Hans-Georg Kopp
- Howard Hughes Medical Institute, Department of Genetic Medicine, Weill Medical College of Cornell University (WMCCU), New York, New York, USA.
Department of Hematology-Oncology, Eberhard-Karls University, Tubingen, Germany.
Divisions of Hematology/Medical Oncology, Medical and Research Service, VA New York Harbor Healthcare System, and Hematology/Medical Oncology, Department of Medicine, WMCCU, New York, New York, USA.
Department of Cell and Developmental Biology, WMCCU, New York, New York, USA.
Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Departments of Biochemistry and Medicine, University of Washington, Seattle, Washington, USA.
Department of Pathology and Laboratory Medicine, WMCCU, New York, New York, USA
| | - Andrea T. Hooper
- Howard Hughes Medical Institute, Department of Genetic Medicine, Weill Medical College of Cornell University (WMCCU), New York, New York, USA.
Department of Hematology-Oncology, Eberhard-Karls University, Tubingen, Germany.
Divisions of Hematology/Medical Oncology, Medical and Research Service, VA New York Harbor Healthcare System, and Hematology/Medical Oncology, Department of Medicine, WMCCU, New York, New York, USA.
Department of Cell and Developmental Biology, WMCCU, New York, New York, USA.
Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Departments of Biochemistry and Medicine, University of Washington, Seattle, Washington, USA.
Department of Pathology and Laboratory Medicine, WMCCU, New York, New York, USA
| | - M. Johan Broekman
- Howard Hughes Medical Institute, Department of Genetic Medicine, Weill Medical College of Cornell University (WMCCU), New York, New York, USA.
Department of Hematology-Oncology, Eberhard-Karls University, Tubingen, Germany.
Divisions of Hematology/Medical Oncology, Medical and Research Service, VA New York Harbor Healthcare System, and Hematology/Medical Oncology, Department of Medicine, WMCCU, New York, New York, USA.
Department of Cell and Developmental Biology, WMCCU, New York, New York, USA.
Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Departments of Biochemistry and Medicine, University of Washington, Seattle, Washington, USA.
Department of Pathology and Laboratory Medicine, WMCCU, New York, New York, USA
| | - Scott T. Avecilla
- Howard Hughes Medical Institute, Department of Genetic Medicine, Weill Medical College of Cornell University (WMCCU), New York, New York, USA.
Department of Hematology-Oncology, Eberhard-Karls University, Tubingen, Germany.
Divisions of Hematology/Medical Oncology, Medical and Research Service, VA New York Harbor Healthcare System, and Hematology/Medical Oncology, Department of Medicine, WMCCU, New York, New York, USA.
Department of Cell and Developmental Biology, WMCCU, New York, New York, USA.
Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Departments of Biochemistry and Medicine, University of Washington, Seattle, Washington, USA.
Department of Pathology and Laboratory Medicine, WMCCU, New York, New York, USA
| | - Isabelle Petit
- Howard Hughes Medical Institute, Department of Genetic Medicine, Weill Medical College of Cornell University (WMCCU), New York, New York, USA.
Department of Hematology-Oncology, Eberhard-Karls University, Tubingen, Germany.
Divisions of Hematology/Medical Oncology, Medical and Research Service, VA New York Harbor Healthcare System, and Hematology/Medical Oncology, Department of Medicine, WMCCU, New York, New York, USA.
Department of Cell and Developmental Biology, WMCCU, New York, New York, USA.
Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Departments of Biochemistry and Medicine, University of Washington, Seattle, Washington, USA.
Department of Pathology and Laboratory Medicine, WMCCU, New York, New York, USA
| | - Min Luo
- Howard Hughes Medical Institute, Department of Genetic Medicine, Weill Medical College of Cornell University (WMCCU), New York, New York, USA.
Department of Hematology-Oncology, Eberhard-Karls University, Tubingen, Germany.
Divisions of Hematology/Medical Oncology, Medical and Research Service, VA New York Harbor Healthcare System, and Hematology/Medical Oncology, Department of Medicine, WMCCU, New York, New York, USA.
Department of Cell and Developmental Biology, WMCCU, New York, New York, USA.
Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Departments of Biochemistry and Medicine, University of Washington, Seattle, Washington, USA.
Department of Pathology and Laboratory Medicine, WMCCU, New York, New York, USA
| | - Till Milde
- Howard Hughes Medical Institute, Department of Genetic Medicine, Weill Medical College of Cornell University (WMCCU), New York, New York, USA.
Department of Hematology-Oncology, Eberhard-Karls University, Tubingen, Germany.
Divisions of Hematology/Medical Oncology, Medical and Research Service, VA New York Harbor Healthcare System, and Hematology/Medical Oncology, Department of Medicine, WMCCU, New York, New York, USA.
Department of Cell and Developmental Biology, WMCCU, New York, New York, USA.
Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Departments of Biochemistry and Medicine, University of Washington, Seattle, Washington, USA.
Department of Pathology and Laboratory Medicine, WMCCU, New York, New York, USA
| | - Carlos A. Ramos
- Howard Hughes Medical Institute, Department of Genetic Medicine, Weill Medical College of Cornell University (WMCCU), New York, New York, USA.
Department of Hematology-Oncology, Eberhard-Karls University, Tubingen, Germany.
Divisions of Hematology/Medical Oncology, Medical and Research Service, VA New York Harbor Healthcare System, and Hematology/Medical Oncology, Department of Medicine, WMCCU, New York, New York, USA.
Department of Cell and Developmental Biology, WMCCU, New York, New York, USA.
Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Departments of Biochemistry and Medicine, University of Washington, Seattle, Washington, USA.
Department of Pathology and Laboratory Medicine, WMCCU, New York, New York, USA
| | - Fan Zhang
- Howard Hughes Medical Institute, Department of Genetic Medicine, Weill Medical College of Cornell University (WMCCU), New York, New York, USA.
Department of Hematology-Oncology, Eberhard-Karls University, Tubingen, Germany.
Divisions of Hematology/Medical Oncology, Medical and Research Service, VA New York Harbor Healthcare System, and Hematology/Medical Oncology, Department of Medicine, WMCCU, New York, New York, USA.
Department of Cell and Developmental Biology, WMCCU, New York, New York, USA.
Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Departments of Biochemistry and Medicine, University of Washington, Seattle, Washington, USA.
Department of Pathology and Laboratory Medicine, WMCCU, New York, New York, USA
| | - Tabitha Kopp
- Howard Hughes Medical Institute, Department of Genetic Medicine, Weill Medical College of Cornell University (WMCCU), New York, New York, USA.
Department of Hematology-Oncology, Eberhard-Karls University, Tubingen, Germany.
Divisions of Hematology/Medical Oncology, Medical and Research Service, VA New York Harbor Healthcare System, and Hematology/Medical Oncology, Department of Medicine, WMCCU, New York, New York, USA.
Department of Cell and Developmental Biology, WMCCU, New York, New York, USA.
Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Departments of Biochemistry and Medicine, University of Washington, Seattle, Washington, USA.
Department of Pathology and Laboratory Medicine, WMCCU, New York, New York, USA
| | - Paul Bornstein
- Howard Hughes Medical Institute, Department of Genetic Medicine, Weill Medical College of Cornell University (WMCCU), New York, New York, USA.
Department of Hematology-Oncology, Eberhard-Karls University, Tubingen, Germany.
Divisions of Hematology/Medical Oncology, Medical and Research Service, VA New York Harbor Healthcare System, and Hematology/Medical Oncology, Department of Medicine, WMCCU, New York, New York, USA.
Department of Cell and Developmental Biology, WMCCU, New York, New York, USA.
Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Departments of Biochemistry and Medicine, University of Washington, Seattle, Washington, USA.
Department of Pathology and Laboratory Medicine, WMCCU, New York, New York, USA
| | - David K. Jin
- Howard Hughes Medical Institute, Department of Genetic Medicine, Weill Medical College of Cornell University (WMCCU), New York, New York, USA.
Department of Hematology-Oncology, Eberhard-Karls University, Tubingen, Germany.
Divisions of Hematology/Medical Oncology, Medical and Research Service, VA New York Harbor Healthcare System, and Hematology/Medical Oncology, Department of Medicine, WMCCU, New York, New York, USA.
Department of Cell and Developmental Biology, WMCCU, New York, New York, USA.
Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Departments of Biochemistry and Medicine, University of Washington, Seattle, Washington, USA.
Department of Pathology and Laboratory Medicine, WMCCU, New York, New York, USA
| | - Aaron J. Marcus
- Howard Hughes Medical Institute, Department of Genetic Medicine, Weill Medical College of Cornell University (WMCCU), New York, New York, USA.
Department of Hematology-Oncology, Eberhard-Karls University, Tubingen, Germany.
Divisions of Hematology/Medical Oncology, Medical and Research Service, VA New York Harbor Healthcare System, and Hematology/Medical Oncology, Department of Medicine, WMCCU, New York, New York, USA.
Department of Cell and Developmental Biology, WMCCU, New York, New York, USA.
Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Departments of Biochemistry and Medicine, University of Washington, Seattle, Washington, USA.
Department of Pathology and Laboratory Medicine, WMCCU, New York, New York, USA
| | - Shahin Rafii
- Howard Hughes Medical Institute, Department of Genetic Medicine, Weill Medical College of Cornell University (WMCCU), New York, New York, USA.
Department of Hematology-Oncology, Eberhard-Karls University, Tubingen, Germany.
Divisions of Hematology/Medical Oncology, Medical and Research Service, VA New York Harbor Healthcare System, and Hematology/Medical Oncology, Department of Medicine, WMCCU, New York, New York, USA.
Department of Cell and Developmental Biology, WMCCU, New York, New York, USA.
Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, USA.
Departments of Biochemistry and Medicine, University of Washington, Seattle, Washington, USA.
Department of Pathology and Laboratory Medicine, WMCCU, New York, New York, USA
| |
Collapse
|
36
|
|
37
|
CD39, NTPDase 1, is attached to the plasma membrane by two transmembrane domains. Why? Purinergic Signal 2006; 2:391-8. [PMID: 18404478 PMCID: PMC2254477 DOI: 10.1007/s11302-005-5907-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2005] [Revised: 12/14/2005] [Accepted: 12/14/2005] [Indexed: 11/20/2022] Open
Abstract
Since the identification of CD39 and other members of the e-NTPDase (ecto-nucleoside triphosphate diphosphohydrolase) family as the primary enzymes responsible for cell surface nucleotide hydrolysis, one of their most intriguing features has been their unusual topology. The active site lies in the large extracellular region, but instead of being anchored in the membrane by a single transmembrane domain or lipid link like other ectoenzymes, CD39 has two transmembrane domains, one at each end. In this review we discuss evidence that the structure and dynamics of the transmembrane helices are intricately connected to enzymatic function. Removal of either or both transmembrane domains or disruption of their native state by detergent solubilization reduces activity by 90%, indicating that native function requires both transmembrane domains to be present and in the membrane. Enzymatic and mutational analysis of the native and truncated forms has shown that the active site can exist in distinct functional states characterized by different total activities, substrate specificities, hydrolysis mechanisms, and intermediate ADP release during ATP hydrolysis, depending on the state of the transmembrane domains. Disulfide crosslinking of cysteines introduced within the transmembrane helices revealed that they interact within and between molecules, in particular near the extracellular domain, and that activity depends on their organization. Both helices exhibit a high degree of rotational mobility, and the ability to undergo dynamic motions is required for activity and regulated by substrate binding. Recent reports suggest that membrane composition can regulate NTPDase activity. We propose that mechanical bilayer properties, potentially elasticity, might regulate CD39 by altering the balance between stability and mobility of its transmembrane domains.
Collapse
|
38
|
Haller CA, Cui W, Wen J, Robson SC, Chaikof EL. Reconstitution of CD39 in liposomes amplifies nucleoside triphosphate diphosphohydrolase activity and restores thromboregulatory properties. J Vasc Surg 2006; 43:816-23. [PMID: 16616242 DOI: 10.1016/j.jvs.2005.11.057] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2005] [Accepted: 11/26/2005] [Indexed: 10/24/2022]
Abstract
BACKGROUND CD39 (nucleoside triphosphate diphosphohydrolase [NTPDase-1]) expressed on the luminal surface of endothelial cells rapidly metabolizes extracellular adenosine triphosphate (ATP) and adenosine diphosphate (ADP) to adenosine monophosphate (AMP), and abrogates platelet reactivity. Optimization of CD39 enzymatic activity appears dependent upon the expression of both transmembrane domains within plasma membranes. Thus, motivation exists to examine therapeutic antiplatelet formulations that consist of liposomal CD39. METHODS Full-length human CD39 was produced by using a yeast expression system, purified, and reconstituted within lipid vesicles. The catalytic efficiency (kcat/Km) of CD39-mediated phosphohydrolysis of ADP and ATP was determined both for detergent-solubilized and protein-reconstituted CD39 within lipid membranes. The capacity of CD39-containing lipid vesicles to inhibit platelet activation induced by ADP, collagen, or thrombin was determined in vitro by platelet aggregometry. A murine model of thromboplastin-induced thromboembolism was used to determine the effectiveness of intravenous liposomal CD39 in limiting platelet consumption and mortality. RESULTS Reconstitution of human CD39 in lipid vesicles was associated with a decrease in Km of nearly an order of magnitude over the detergent-solubilized form. There was a concomitant increase in both ADPase and ATPase catalytic efficiencies (kcat/Km ADPase: sol CD39: 2.7 x 10(6) vs liposomal CD39: 1.4 x 10(7) min/ M; kcat/Km ATPase: sol CD39: 7.2 x 10(6) vs liposomal CD39: 2.0 x10(7) min/M). Furthermore, CD39 lipid vesicles effectively inhibited platelet aggregation when activated by ADP, collagen, or thrombin, and also promoted platelet disaggregation (60.4% +/- 6.1%). Treatment with CD39 lipid vesicles preserved platelet counts after thromboplastin injection (pretreatment, 906.8 +/- 42.9 platelets/microm3; empty vesicles, 278.6 +/- 34.8 platelets/microm3; CD39 vesicles, 563.6 +/- 42.2 platelets/microm3; n = 10 mice/test group; P < .0001). In parallel survival studies, liposomal CD39 reduced mortality from 73% to 33% (P < or = .05; n = 12 mice/experimental test group, n = 15 mice/control test group). CONCLUSIONS Incorporation of solubilized CD39 into a lipid bilayer restores enzyme activity and optimizes thromboregulatory potential. Treatment with CD39 in liposomal formulations decreased mortality in a murine model of thromboplastin-induced thromboembolism by limiting intravascular platelet aggregation and thrombosis.
Collapse
|
39
|
Massé K, Eason R, Bhamra S, Dale N, Jones EA. Comparative genomic and expression analysis of the conserved NTPDase gene family in Xenopus. Genomics 2006; 87:366-81. [PMID: 16380227 DOI: 10.1016/j.ygeno.2005.11.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2005] [Revised: 10/17/2005] [Accepted: 11/09/2005] [Indexed: 10/25/2022]
Abstract
The purines, ATP and adenosine, are important signaling molecules in the nervous system. ATP is sequentially degraded to adenosine by the ectonucleotidase proteins. The NTPDase (or CD39) family is a subfamily of these enzymes, which consists of nine members in mammals. In Xenopus embryos, we have shown that ATP, and its antagonist adenosine, regulate the rundown of swimming and we therefore proposed that ectonucleotidase proteins are key regulators of locomotor activity. Here, we report the cloning of all nine members of the NTPDase family in Xenopus laevis and Xenopus tropicalis. Our phylogenetic analysis shows that this family is highly conserved between the frog species and also during vertebrate evolution. In the adult frog, NTPDase genes are broadly expressed. During development, all NTPDase genes, except for NTPDase8, are expressed and display a distinct specific expression pattern, suggesting potentially different functions of these proteins during embryogenesis of X. laevis.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens, CD/genetics
- Apyrase/genetics
- Cloning, Molecular
- DNA, Complementary/chemistry
- DNA, Complementary/genetics
- DNA, Complementary/isolation & purification
- Embryo, Nonmammalian/embryology
- Embryo, Nonmammalian/metabolism
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Developmental
- Genomics/methods
- In Situ Hybridization
- Isoenzymes/genetics
- Male
- Molecular Sequence Data
- Multigene Family/genetics
- Phylogeny
- Sequence Alignment
- Sequence Analysis, DNA
- Sequence Homology, Amino Acid
- Time Factors
- Xenopus/embryology
- Xenopus/genetics
- Xenopus/growth & development
- Xenopus Proteins/genetics
- Xenopus laevis/embryology
- Xenopus laevis/genetics
- Xenopus laevis/growth & development
Collapse
Affiliation(s)
- Karine Massé
- Department of Biological Sciences, Warwick University, Coventry, CV47AL, UK
| | | | | | | | | |
Collapse
|
40
|
Buergler JM, Maliszewski CR, Broekman MJ, Kaluza GL, Schulz DG, Marcus AJ, Raizner AE, Kleiman NS, Ali NM. Effects of SolCD39, a novel inhibitor of Platelet Aggregation, on Platelet Deposition and Aggregation after PTCA in a Porcine Model. J Thromb Thrombolysis 2005; 19:115-22. [PMID: 16052302 DOI: 10.1007/s11239-005-1381-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
INTRODUCTION This study evaluated CD39 in a porcine model of balloon angioplasty and in plasma of patients undergoing percutaneous intervention. CD39 (E-NTPDase1), is the endothelial ecto-ADPase inhibiting platelet function via hydrolysis of released platelet ADP. METHODS AND RESULTS A recombinant soluble form of CD39 (solCD39) given intravenously to pigs had an elimination half life of 5--7 days, increased the bleeding time to an extent similar to aspirin, and inhibits platelet aggregation by>90%. Platelet counts and clot retraction remained normal following solCD39 administration. In a pig model of acute coronary balloon injury, solCD39 resulted in non-statistically significant decreases in platelet (7.7+/-1.4 versus 11.7+/- 3.4) and fibrin (3.5+/- 0.4 versus 4.2+/- 0.7) deposition ratios. Adding ex vivo to human platelet rich plasma (PRP) solCD39 produced nearly 100% inhibition of ADP-induced platelet aggregation. A dose-response effect of solCD39 on platelet aggregation induced by collagen or a thrombin receptor activating peptide (TRAP(SFLLRN)) was noted in PRP obtained from volunteers and patients receiving aspirin, clopidogrel or ticlopidine. SolCD39 also provided additional and complete inhibition of TRAP-induced platelet aggregation in PRP from patients who had received abciximab, aspirin and clopidogrel. CONCLUSIONS SolCD39, a novel inhibitor of platelet activation and recruitment with a relatively long half-life appears to be well tolerated and is a potent inhibitor of ADP-, collagen-, or TRAP-induced platelet activation. Its potential use in percutaneous coronary intervention requires further study. ABBREVIATED ABSTRACT: E-NTPDase1/CD39 is the endothelial ecto-ADPase responsible for inhibition of platelet function. A recombinant soluble form (solCD39) had an elimination half life of 5-7 days in pigs, elevated bleeding times similar to aspirin, did not affect clot retraction, and inhibited platelet aggregation by > 90%. When combined with standard heparin therapy in a pig model of acute coronary balloon injury, solCD39 resulted in a trend toward a decrease in platelet and fibrin deposition. SolCD39 added ex vivo to human platelet rich plasma yielded nearly 100% inhibition of ADP-induced platelet aggregation and provided further inhibition when combined with standard therapy.
Collapse
Affiliation(s)
- John M Buergler
- Section of Cardiology, Department of Medicine, Baylor College of Medicine, The Methodist DeBakey Heart Center, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Maoz T, Koren R, Ben-Ari I, Kleinberger T. YND1 interacts with CDC55 and is a novel mediator of E4orf4-induced toxicity. J Biol Chem 2005; 280:41270-7. [PMID: 16227198 DOI: 10.1074/jbc.m507281200] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Adenovirus E4orf4 (early region 4 open reading frame 4) protein induces protein phosphatase 2A-dependent non-classical apoptosis in mammalian cells and irreversible growth arrest in Saccharomyces cerevisiae. Oncogenic transformation sensitizes cells to E4orf4-induced cell death. To uncover additional components of the E4orf4 network required for induction of its unique mode of apoptosis, we used yeast genetics to select gene deletions conferring resistance to E4orf4. Deletion of YND1, encoding a yeast Golgi apyrase, conferred partial resistance to E4orf4. However, Ynd1p apyrase activity was not required for E4orf4-induced toxicity. Ynd1p and Cdc55p, the yeast protein phosphatase 2A-B subunit, contributed additively to E4orf4-induced toxicity. Furthermore, concomitant overexpression of one and deletion of the other was detrimental to yeast growth, demonstrating a functional interaction between the two proteins. YND1 and CDC55 also interacted genetically with CDC20 and CDH1/HCT1, encoding activating subunits of the anaphase-promoting complex/cyclosome. In addition to their functional interaction, Ynd1p and Cdc55p interacted physically, and this interaction was disrupted by E4orf4, which remained associated with both proteins. The results suggested that Ynd1p and Cdc55p share a common downstream target whose balanced modulation by the two E4orf4 partners is crucial to viability. Disruption of this balance by E4orf4 may lead to cell death. NTPDase-4/Lalp70/UDPase, the closest mammalian homologue of Ynd1p, associated with E4orf4 in mammalian cells, suggesting that the results in yeast are relevant to the mammalian system.
Collapse
Affiliation(s)
- Tsofnat Maoz
- The Gonda Center of Molecular Microbiology and The Rappaport Family Institute for Research in the Medical Sciences, Faculty of Medicine, Technion-Israel Institute of Technology, Bat Galim, Haifa, 31096, Israel
| | | | | | | |
Collapse
|
42
|
El-Omar MM, Islam N, Broekman MJ, Drosopoulos JHF, Roa DC, Lorin JD, Sedlis SP, Olson KE, Pulte ED, Marcus AJ. The ratio of ADP- to ATP-ectonucleotidase activity is reduced in patients with coronary artery disease. Thromb Res 2005; 116:199-206. [PMID: 15935828 DOI: 10.1016/j.thromres.2004.11.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2004] [Revised: 11/02/2004] [Accepted: 11/28/2004] [Indexed: 11/16/2022]
Abstract
INTRODUCTION CD39 (NTPDase1), an endothelial cell membrane glycoprotein, is the predominant ATP diphosphohydrolase (ATPDase) in vascular endothelium. It hydrolyses both triphosphonucleosides and diphosphonucleosides at comparable rates, thus terminating platelet aggregation and recruitment responses to ADP and other platelet agonists. This occurs even when nitric oxide (NO) formation and prostacyclin production are inhibited. Thus, CD39 represents the main control system for platelet reactivity. Reduced or deficient local ecto-nucleotidase activity may predispose to development of vascular disease. Based on data in animal models and in vitro, CD39 constitutes a new therapeutic modality for vascular disease with a novel and unique mode of action. MATERIALS AND METHODS Lymphocytes were isolated from 46 patients with angiographically proven coronary artery disease (CAD) as well as from matched healthy control subjects. Ectonucleotidase ADPase and ATPase activities (prototypical for the ATPDase activity of endothelial cells) were measured using established radio-TLC procedures. RESULTS AND DISCUSSION In the patients, a decreased ratio of ADPase to ATPase activities (from 1.26 to 1.04) was observed despite increases in both ADPase and ATPase activities. Coronary artery disease was the only independent predictor of a difference in the ADPase/ATPase activity ratio by multivariate linear regression analysis (P=0.0035). This altered ADPase/ATPase activity ratio in patients may represent a reduction in endogenous defense systems against platelet-driven thrombotic events. These data may identify a population of patients with excessive platelet reactivity in their circulation. Increased generation of prothrombotic ADP in these patients implies a potential benefit from therapeutic intervention with soluble forms of CD39.
Collapse
Affiliation(s)
- Magdi M El-Omar
- Department of Medicine-Cardiology, New York University Medical School, New York, NY, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Marcus AJ, Broekman MJ, Drosopoulos JHF, Islam N, Pinsky DJ, Sesti C, Levi R. Heterologous cell-cell interactions: thromboregulation, cerebroprotection and cardioprotection by CD39 (NTPDase-1). J Thromb Haemost 2003; 1:2497-509. [PMID: 14675084 DOI: 10.1111/j.1538-7836.2003.00479.x] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Blood platelets maintain vascular integrity and promote primary and secondary hemostasis following interruption of vessel continuity. Biochemical or physical damage to the coronary, carotid or peripheral arteries is followed by excessive platelet activation and recruitment culminating in vascular occlusion and tissue ischemia. Currently inadequate therapeutic approaches to stroke and coronary artery disease are a public health issue. Following our demonstration of neutrophil leukotriene production from arachidonate released from activated aspirin-treated platelets, we studied interactions between platelets and other blood cells, leading to concepts of transcellular metabolism and thromboregulation. Thrombosis has a proinflammatory component whereby biologically active substances are synthesized by interactions between different cell types that could not individually synthesize the product(s). Endothelial cells control platelet reactivity via three biochemical systems-autacoids leading to production of prostacyclin and nitric oxide, and endothelial ecto-ADPase/CD39/NTPDase-1. The autacoids are fluid-phase reactants, not produced by tissues in the basal state. They are only synthesized intracellularly and released upon interactions of cells with an agonist. When released, autacoids exert fleeting actions in the immediate milieu, and are rapidly inactivated. CD39 is an integral component of the endothelial cell surface and is substrate-activated. It maintains vascular fluidity in the complete absence of prostacyclin and nitric oxide, indicating that they are ancillary components of hemostasis. Therapeutic implications for the autacoids have not been compelling because of their transient, local and fleeting action, and limited potency. Conversely, CD39, acting solely on the platelet releasate, is efficacious in three different animal models. It metabolically neutralizes a prothrombotic platelet releasate via deletion of ADP--the major recruiting agent responsible for formation of an occlusive thrombus. In addition, solCD39 reduced ATP- and ischemia-induced norepinephrine release in the heart. This reduction can prevent fatal arrhythmia. Moreover, solCD39 ameliorated the sequelae of stroke in CD39 null mice. CD39 represents the next generation of cardioprotective and cerebroprotective molecules.
Collapse
Affiliation(s)
- A J Marcus
- Department of Medicine, Weill Medical College of Cornell University, and Medical Service/Hematology-Oncology, VA New York Harbor Healthcare System, New York, NY 10010, USA.
| | | | | | | | | | | | | |
Collapse
|
44
|
Sesti C, Koyama M, Broekman MJ, Marcus AJ, Levi R. Ectonucleotidase in sympathetic nerve endings modulates ATP and norepinephrine exocytosis in myocardial ischemia. J Pharmacol Exp Ther 2003; 306:238-44. [PMID: 12676879 DOI: 10.1124/jpet.103.049874] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We recently reported that ATP, coreleased with norepinephrine (NE) from cardiac sympathetic nerves, increases NE exocytosis via a positive feedback mechanism. A neuronal ectonucleotidase (E-NTPDase) metabolizes the released ATP, decreasing NE exocytosis. Excessive NE release in myocardial ischemia exacerbates cardiac dysfunction. Thus, we studied whether the ATP-mediated autocrine amplification of NE release is operative in ischemia and, if so, whether it can be modulated by E-NTPDase and its recombinant equivalent, solCD39. Isolated, guinea pig hearts underwent 10- or 20-min ischemic episodes, wherein NE was released by exocytosis and reversal of the NE transporter, respectively. Furthermore, to restrict the role of E-NTPDase to transmitter ATP, sympathetic nerve endings were isolated (cardiac synaptosomes) and subjected to increasing periods of ischemia. Availability of released ATP at the nerve terminals was either increased via E-NTPDase inhibition or diminished by enhancing ATP hydrolysis with solCD39. P2X receptor blockade with PPADS was used to attenuate the effects of released ATP. We found that, in short-term ischemia (but, as anticipated, not in protracted ischemia, where NE release is carrier-mediated), ATP exocytosis was linearly correlated with that of NE. This indicates that by limiting the availability of ATP at sympathetic terminals, E-NTPDase effectively attenuates NE exocytosis in myocardial ischemia. Our findings suggest a key role for neuronal E-NTPDase in the control of adrenergic function in the ischemic heart. Because excessive NE release is an established cause of dysfunction in ischemic heart disease, solCD39 may offer a novel therapeutic approach to myocardial ischemia and its consequences.
Collapse
Affiliation(s)
- Casilde Sesti
- Department of Pharmacology, Room LC419, 1300 York Ave., Weill Medical College of Cornell University, New York, NY 10021, USA
| | | | | | | | | |
Collapse
|
45
|
Marcus AJ, Broekman MJ, Drosopoulos JHF, Islam N, Pinsky DJ, Sesti C, Levi R. Metabolic control of excessive extracellular nucleotide accumulation by CD39/ecto-nucleotidase-1: implications for ischemic vascular diseases. J Pharmacol Exp Ther 2003; 305:9-16. [PMID: 12649347 DOI: 10.1124/jpet.102.043729] [Citation(s) in RCA: 105] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Platelets are responsible for maintaining vascular integrity. In thrombocytopenic states, vascular permeability and fragility increase, presumably due to the absence of this platelet function. Chemical or physical injury to a blood vessel induces platelet activation and platelet recruitment. This is beneficial for the arrest of bleeding (hemostasis), but when an atherosclerotic plaque is ulcerated or fissured, it becomes an agonist for vascular occlusion (thrombosis). Experiments in the late 1980s cumulatively indicated that endothelial cell CD39-an ecto-ADPase-reduced platelet reactivity to most agonists, even in the absence of prostacyclin or nitric oxide. As discussed herein, CD39 rapidly and preferentially metabolizes ATP and ADP released from activated platelets to AMP, thereby drastically reducing or even abolishing platelet aggregation and recruitment. Since ADP is the final common agonist for platelet recruitment and thrombus formation, this finding highlights the significance of CD39. A recombinant, soluble form of human CD39, solCD39, has enzymatic and biological properties identical to the full-length form of the molecule and strongly inhibits human platelet aggregation induced by ADP, collagen, arachidonate, or TRAP (thrombin receptor agonist peptide). In sympathetic nerve endings isolated from guinea pig hearts, where neuronal ATP enhances norepinephrine exocytosis, solCD39 markedly attenuated norepinephrine release. This suggests that NTPDase (nucleoside triphosphate diphosphohydrolase) could exert a cardioprotective action by reducing ATP-mediated norepinephrine release, thereby offering a novel therapeutic approach to myocardial ischemia and its consequences. In a murine model of stroke, driven by excessive platelet recruitment, solCD39 reduced the sequelae of stroke, without an increase in intracerebral hemorrhage. CD39 null mice, generated by deletion of apyrase-conserved regions 2 to 4, exhibited a decrease in postischemic perfusion and an increase in cerebral infarct volume when compared with controls. "Reconstitution" of CD39 null mice with solCD39 reversed these changes. We hypothesize that solCD39 has potential as a novel therapeutic agent for thrombotic diatheses.
Collapse
Affiliation(s)
- Aaron J Marcus
- Department of Medicine, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York 10010, USA.
| | | | | | | | | | | | | |
Collapse
|
46
|
Birk AV, Broekman MJ, Gladek EM, Robertson HD, Drosopoulos JHF, Marcus AJ, Szeto HH. Role of extracellular ATP metabolism in regulation of platelet reactivity. ACTA ACUST UNITED AC 2002; 140:166-75. [PMID: 12271274 DOI: 10.1067/mlc.2002.126719] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Extracellular adenosine triphosphate (ATP) regulates platelet reactivity by way of direct action on platelet purinergic receptors or by hydrolysis to adenosine diphosphate (ADP). Subsequent metabolism of ATP and ADP to adenosine monophosphate (AMP) and adenosine inhibits platelet aggregation. Endothelial cell membrane-bound ecto-ATP/ADPase (CD39, E-NTPDase1) is thought to be the main regulator of platelet responsiveness. However, the findings in studies of CD39-knockout mice imply that nucleotidase(s) in plasma regulates circulating adenine nucleotides levels. Understanding extracellular ATP metabolism by CD39 and plasma nucleotidases is therefore important. In this study, alpha-phosphorus 32- and gamma-phosphorus 32-labeled ATP were rapidly metabolized directly to AMP and pyrophosphate in human plasma at pH 7.4, suggesting the presence of pyrophosphatase/phosphodiesterase-like activity. A specific phosphodiesterase substrate, p-nitrophenol-5'-TMP (p-Nph-5'-TMP), was readily hydrolyzed in human plasma. The antiaggregatory action of beta,gamma-methylene-ATP (AMPPCP) (5 micromol/L) was blocked by DMPX, an adenosine-receptor antagonist, suggesting that in plasma, AMPPCP was metabolized to AMP and adenosine. Recombinant soluble CD39 (solCD39) was used to assess the role of CD39 in ATP metabolism. As little as 0.25 microg/mL of solCD39 inhibited ADP-induced platelet aggregation. However, in the presence of ADP-free ATP (10 micromol/L), solCD39 induced platelet aggregation in a dose-dependent manner. Because AMPPCP could not substitute for ATP in solCD39-stimulated platelet aggregation, it is likely that ADP formation from ATP was required. Endogenous CD39 may thus have a hemostatic function by promoting ADP formation from released ATP, in addition to its antiaggregatory properties. A plasma nucleotidase hydrolyzes ATP directly to AMP. This prevents ADP accumulation and generates adenosine, a potent, locally acting inhibitor of platelet reactivity. The presence of both endothelial CD39 and plasma nucleotidase appears to be important in the maintenance of normal hemostasis and prevention of excessive platelet responsiveness.
Collapse
Affiliation(s)
- Alex V Birk
- Department of Pharmacology, Weill Medical College of Cornell University, New York, NY 10021, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Drosopoulos JHF. Roles of Asp54 and Asp213 in Ca2+ utilization by soluble human CD39/ecto-nucleotidase. Arch Biochem Biophys 2002; 406:85-95. [PMID: 12234494 DOI: 10.1016/s0003-9861(02)00414-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Soluble human CD39 (solCD39) rapidly metabolizes nucleotides, especially ADP released from activated platelets, thereby inhibiting further platelet activation and recruitment. Using alanine substitution mutagenesis, we established a functional role for aspartates D54 and D213 in solCD39. Kinetic analyses of D54A and D213A indicated decreased K(m)s of the mutants, compared to wild type, for the cofactor calcium and for the substrates ADP and ATP. These decreases in calcium and nucleotide affinity of the mutants were accompanied by increases in their rate of catalysis. The decreased affinity of the mutants for calcium was responsible for their diminished ability to reverse platelet aggregation in plasma anticoagulated with citrate, a known calcium chelator. Their ADPase activity in the presence of citrated plasma was also decreased, although this could be overcome with excess calcium. Thus, aspartates 54 and 213 are involved in calcium utilization and potentially involved in cation coordination with substrate in the catalytic pocket of solCD39.
Collapse
Affiliation(s)
- Joan H F Drosopoulos
- Research Service, Thrombosis Research Laboratory, VA New York Harbor Healthcare System, New York, NY 10010-5050, USA.
| |
Collapse
|
48
|
Smith TM, Hicks-Berger CA, Kim S, Kirley TL. Cloning, expression, and characterization of a soluble calcium-activated nucleotidase, a human enzyme belonging to a new family of extracellular nucleotidases. Arch Biochem Biophys 2002; 406:105-15. [PMID: 12234496 DOI: 10.1016/s0003-9861(02)00420-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The salivary apyrases of blood-feeding arthropods are nucleotide-hydrolyzing enzymes implicated in the inhibition of host platelet aggregation through the hydrolysis of extracellular adenosine diphosphate. A human cDNA homologous to the apyrase cDNA of the blood-feeding bed bug was identified, revealing an open reading frame encoding a 371-amino acid protein. A cleavable signal peptide generates a secreted protein of 333 residues with a predicted core molecular mass of 37,193 Da. Expression in COS-1 cells produced a secreted apyrase in the cell media. The ADPase and ATPase activities were dependent upon calcium, with a pH optimum between pH 6.2 and 7.2. Interestingly, the preferred substrate was not ADP, as might be expected for an enzyme modulating platelet aggregation, but rather UDP, followed by GDP, UTP, GTP, ADP, and ATP. The nucleotidase did not hydrolyze nucleoside monophosphates. Size-exclusion chromatography and Western blot analysis revealed a molecular mass of approximately 34-37 kDa. Treatment of the enzyme with peptide N-glycosidase F indicated that the protein is glycosylated. Northern analysis identified the transcript in a range of human tissues, including testis, placenta, prostate, and lung. No traditional apyrase-conserved regions or nucleotide-binding domains were identified in this human enzyme, indicating membership in a new family of extracellular nucleotidases.
Collapse
Affiliation(s)
- Thomas M Smith
- Department of Inflammation Biology, Wyeth Research, 200 CambridgePark Drive, Cambridge, MA 02140, USA.
| | | | | | | |
Collapse
|
49
|
Abstract
Hemostasis involves a carefully regulated balance between circulating and endothelium-derived prothrombotic and antithrombotic factors. The unstable or vulnerable plaque facilitates thrombosis, clinically manifest as an acute coronary syndrome (ACS), by creating an environment that favors thrombus formation over prevention of lysis. Endothelial cell dysfunction is integral to both the development of the atherosclerotic lesion as well as its destabilization. The transformation of a stable plaque to an unstable one involves complex interactions among T lymphocytes, macrophages, endothelial cells, and smooth muscle cells. Degradation of the fibrous cap of the atherosclerotic lesion as well as the overexpression of prothrombotic and underexpression of antithrombotic factors by cells within the plaque precede thrombus formation. Accordingly, pharmacological interventions for the treatment of ACS are directed against the initiation and propagation of thrombosis, as well as toward improvement of endothelial function.
Collapse
Affiliation(s)
- Frederick L Ruberg
- Evans Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118-2526, USA
| | | | | |
Collapse
|
50
|
Sesti C, Broekman MJ, Drosopoulos JHF, Islam N, Marcus AJ, Levi R. EctoNucleotidase in cardiac sympathetic nerve endings modulates ATP-mediated feedback of norepinephrine release. J Pharmacol Exp Ther 2002; 300:605-11. [PMID: 11805223 DOI: 10.1124/jpet.300.2.605] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
ATP, coreleased with norepinephrine, affects adrenergic transmission by acting on purinoceptors at sympathetic nerve endings. Ectonucleotidases terminate the actions of ATP. Previously, we had preliminary evidence for ectonucleotidase activity in cardiac sympathetic nerve terminals. Therefore, we investigated whether this ectonucleotidase might influence norepinephrine release in the heart. Sympathetic nerve endings isolated from guinea pig heart (cardiac synaptosomes) were rich in Ca(2+)-dependent ectonucleotidase activity, as measured by metabolism of exogenously added radiolabeled ATP or ADP. By its inhibitor profile, ectonucleotidase resembled ectonucleoside triphosphate diphosphohydrolase 1 (E-NTPDase1). Exogenous ATP elicited concentration-dependent norepinephrine release from cardiac synaptosomes (EC(50) 0.96 microM). This release was antagonized by the P2X receptor antagonist pyridoxalphosphate-6-azophenyl-2',4'-disulfonic acid (PPADS) (10 microM) and potentiated by the P2Y receptor antagonist 2'-deoxy-N(6)-methyladenosine-3',5'-diphosphate (MRS 2179) (30 nM). Norepinephrine release promoted by ATP was also potentiated by the nucleotidase inhibitor 6-N,N-diethyl-beta-gamma-dibromomethylene-D-adenosine-5'-triphosphate (ARL67156) (30 microM) and blocked by a recombinant, soluble form of human E-NTPDase1 (solCD39). In contrast, ARL67156 had no effect on norepinephrine release induced by the nonhydrolyzable analog, alpha, beta-methyleneadenosine-5'-triphosphate (alpha,beta-MeATP). Depolarization of cardiac synaptosomes with K(+) elicited release of endogenous norepinephrine. This was attenuated by PPADS and solCD39 and potentiated by MRS 2179 and ARL67156. Importantly, our results demonstrate that facilitation of ATP-induced norepinephrine release from cardiac sympathetic nerves is a composite of two autocrine components: positive, mediated by P2X receptors, and negative, mediated by P2Y receptors. Modulation of norepinephrine release by coreleased ATP is terminated by endogenous as well as exogenous ectonucleotidase. We propose that ectonucleotidase control of norepinephrine release should provide cardiac protection in hyperadrenergic states such as myocardial ischemia.
Collapse
Affiliation(s)
- Casilde Sesti
- Department of Pharmacology, Weill Medical College of Cornell University, New York, New York 10021, USA
| | | | | | | | | | | |
Collapse
|