1
|
Ferrucci V, Lomada S, Wieland T, Zollo M. PRUNE1 and NME/NDPK family proteins influence energy metabolism and signaling in cancer metastases. Cancer Metastasis Rev 2024; 43:755-775. [PMID: 38180572 PMCID: PMC11156750 DOI: 10.1007/s10555-023-10165-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/19/2023] [Indexed: 01/06/2024]
Abstract
We describe here the molecular basis of the complex formation of PRUNE1 with the tumor metastasis suppressors NME1 and NME2, two isoforms appertaining to the nucleoside diphosphate kinase (NDPK) enzyme family, and how this complex regulates signaling the immune system and energy metabolism, thereby shaping the tumor microenvironment (TME). Disrupting the interaction between NME1/2 and PRUNE1, as suggested, holds the potential to be an excellent therapeutic target for the treatment of cancer and the inhibition of metastasis dissemination. Furthermore, we postulate an interaction and regulation of the other Class I NME proteins, NME3 and NME4 proteins, with PRUNE1 and discuss potential functions. Class I NME1-4 proteins are NTP/NDP transphosphorylases required for balancing the intracellular pools of nucleotide diphosphates and triphosphates. They regulate different cellular functions by interacting with a large variety of other proteins, and in cancer and metastasis processes, they can exert pro- and anti-oncogenic properties depending on the cellular context. In this review, we therefore additionally discuss general aspects of class1 NME and PRUNE1 molecular structures as well as their posttranslational modifications and subcellular localization. The current knowledge on the contributions of PRUNE1 as well as NME proteins to signaling cascades is summarized with a special regard to cancer and metastasis.
Collapse
Affiliation(s)
- Veronica Ferrucci
- Department of Molecular Medicine and Medical Biotechnology, DMMBM, University of Naples, Federico II, Via Pansini 5, 80131, Naples, Italy
- CEINGE Biotecnologie Avanzate "Franco Salvatore", Via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Santosh Lomada
- Experimental Pharmacology Mannheim, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany
- DZHK, German Center for Cardiovascular Research, Partner Site Heidelberg/Mannheim, 68167, Mannheim, Germany
| | - Thomas Wieland
- Experimental Pharmacology Mannheim, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany.
- DZHK, German Center for Cardiovascular Research, Partner Site Heidelberg/Mannheim, 68167, Mannheim, Germany.
- Medical Faculty Mannheim, Ludolf Krehl-Str. 13-17, 68167, Mannheim, Germany.
| | - Massimo Zollo
- Department of Molecular Medicine and Medical Biotechnology, DMMBM, University of Naples, Federico II, Via Pansini 5, 80131, Naples, Italy.
- CEINGE Biotecnologie Avanzate "Franco Salvatore", Via Gaetano Salvatore 486, 80145, Naples, Italy.
- DAI Medicina di Laboratorio e Trasfusionale, 'AOU' Federico II Policlinico, 80131, Naples, Italy.
| |
Collapse
|
2
|
Abstract
Glucose-induced (physiological) insulin secretion from the islet β-cell involves interplay between cationic (i.e., changes in intracellular calcium) and metabolic (i.e., generation of hydrophobic and hydrophilic second messengers) events. A large body of evidence affirms support for novel regulation, by G proteins, of specific intracellular signaling events, including actin cytoskeletal remodeling, transport of insulin-containing granules to the plasma membrane for fusion, and secretion of insulin into the circulation. This article highlights the following aspects of GPCR-G protein biology of the islet. First, it overviews our current understanding of the identity of a wide variety of G protein regulators and their modulatory roles in GPCR-G protein-effector coupling, which is requisite for optimal β-cell function under physiological conditions. Second, it describes evidence in support of novel, noncanonical, GPCR-independent mechanisms of activation of G proteins in the islet. Third, it highlights the evidence indicating that abnormalities in G protein function lead to islet β-cell dysregulation and demise under the duress of metabolic stress and diabetes. Fourth, it summarizes observations of potential beneficial effects of GPCR agonists in preventing/halting metabolic defects in the islet β-cell under various pathological conditions (e.g., metabolic stress and inflammation). Lastly, it identifies knowledge gaps and potential avenues for future research in this evolving field of translational islet biology. Published 2020. Compr Physiol 10:453-490, 2020.
Collapse
Affiliation(s)
- Anjaneyulu Kowluru
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Center for Translational Research in Diabetes, Biomedical Research Service, John D. Dingell VA Medical Center, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
3
|
Nucleoside Diphosphate Kinase B Contributes to Arrhythmogenesis in Human-Induced Pluripotent Stem Cell-Derived Cardiomyocytes from a Patient with Arrhythmogenic Right Ventricular Cardiomyopathy. J Clin Med 2020; 9:jcm9020486. [PMID: 32050722 PMCID: PMC7073527 DOI: 10.3390/jcm9020486] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 01/30/2020] [Accepted: 02/05/2020] [Indexed: 12/29/2022] Open
Abstract
Background: Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a rare, inheritable cardiac disorder characterized by ventricular tachyarrhythmias, progressive loss of cardiomyocytes with fibrofatty replacement and sudden cardiac death. The exact underlying mechanisms are unclear. Methods: This study investigated the possible roles of nucleoside diphosphate kinase B (NDPK-B) and SK4 channels in the arrhythmogenesis of ARVC by using human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Results: In hiPSC-CMs from a patient with ARVC, the expression levels of NDPK-B and SK4 channels were upregulated, the cell automaticity was increased and the occurrence rate of arrhythmic events was enhanced. Recombinant NDPK-B applied into hiPSC-CMs from either healthy donors or the patient enhanced SK4 channel current (ISK4), cell automaticity and the occurrence of arrhythmic events, whereas protein histidine phosphatase 1 (PHP-1), a counter actor of NDPK-B, prevented the NDPK-B effect. Application of PHP-1 alone or a SK4 channel blocker also reduced cell automaticity and arrhythmic events. Conclusion: This study demonstrated that the elevated NDPK-B expression, via activating SK4 channels, contributes to arrhythmogenesis in ARVC, and hence, NDPK-B may be a potential therapeutic target for treating arrhythmias in patients with ARVC.
Collapse
|
4
|
Romani P, Ignesti M, Gargiulo G, Hsu T, Cavaliere V. Extracellular NME proteins: a player or a bystander? J Transl Med 2018; 98:248-257. [PMID: 29035383 DOI: 10.1038/labinvest.2017.102] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/27/2017] [Accepted: 07/20/2017] [Indexed: 12/14/2022] Open
Abstract
The Nm23/NME gene family has been under intensive study since Nm23H1/NME1 was identified as the first metastasis suppressor. Inverse correlation between the expression levels of NME1/2 and prognosis has indeed been demonstrated in different tumor cohorts. Interestingly, the presence of NME proteins in the extracellular environment in normal and tumoral conditions has also been noted. In many reported cases, however, these extracellular NME proteins exhibit anti-differentiation or oncogenic functions, contradicting their canonical anti-metastatic action. This emerging field thus warrants further investigation. In this review, we summarize the current understanding of extracellular NME proteins. A role in promoting stem cell pluripotency and inducing development of central nervous system as well as a neuroprotective function of extracellular NME have been suggested. Moreover, a tumor-promoting function of extracellular NME also emerged at least in some tumor cohorts. In this complex scenario, the secretory mechanism through which NME proteins exit cells is far from being understood. Recently, some evidence obtained in the Drosophila and cancer cell line models points to the involvement of Dynamin in controlling the balance between intra- and extracellular levels of NME. Further analyses on extracellular NME will lead to a better understanding of its physiological function and in turn will allow understanding of how its deregulation contributes to carcinogenesis.
Collapse
Affiliation(s)
- Patrizia Romani
- Dipartimento di Farmacia e biotecnologie, Alma Mater Studiorum Università di Bologna, Bologna, Italia
| | - Marilena Ignesti
- Dipartimento di Farmacia e biotecnologie, Alma Mater Studiorum Università di Bologna, Bologna, Italia
| | - Giuseppe Gargiulo
- Dipartimento di Farmacia e biotecnologie, Alma Mater Studiorum Università di Bologna, Bologna, Italia
| | - Tien Hsu
- Boston University School of Medicine, Department of Medicine, Boston, MA, USA.,National Central University, Department of Biomedical Sciences and Technology, Jhongli, Taiwan
| | - Valeria Cavaliere
- Dipartimento di Farmacia e biotecnologie, Alma Mater Studiorum Università di Bologna, Bologna, Italia
| |
Collapse
|
5
|
Abu-Taha IH, Heijman J, Feng Y, Vettel C, Dobrev D, Wieland T. Regulation of heterotrimeric G-protein signaling by NDPK/NME proteins and caveolins: an update. J Transl Med 2018; 98:190-197. [PMID: 29035382 DOI: 10.1038/labinvest.2017.103] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 07/17/2017] [Accepted: 07/31/2017] [Indexed: 12/14/2022] Open
Abstract
Heterotrimeric G proteins are pivotal mediators of cellular signal transduction in eukaryotic cells and abnormal G-protein signaling plays an important role in numerous diseases. During the last two decades it has become evident that the activation status of heterotrimeric G proteins is both highly localized and strongly regulated by a number of factors, including a receptor-independent activation pathway of heterotrimeric G proteins that does not involve the classical GDP/GTP exchange and relies on nucleoside diphosphate kinases (NDPKs). NDPKs are NTP/NDP transphosphorylases encoded by the nme/nm23 genes that are involved in a variety of cellular events such as proliferation, migration, and apoptosis. They therefore contribute, for example, to tumor metastasis, angiogenesis, retinopathy, and heart failure. Interestingly, NDPKs are translocated and/or upregulated in human heart failure. Here we describe recent advances in the current understanding of NDPK functions and how they have an impact on local regulation of G-protein signaling.
Collapse
Affiliation(s)
- Issam H Abu-Taha
- Institute of Pharmacology, West-German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Jordi Heijman
- Department of Cardiology, CARIM School for Cardiovascular Disease, Maastricht University, Maastricht, The Netherlands
| | - Yuxi Feng
- Institute of Experimental and Clinical Pharmacology and Toxicology, Mannheim Medical Faculty, Heidelberg University, Mannheim, Germany
| | - Christiane Vettel
- Institute of Experimental and Clinical Pharmacology and Toxicology, Mannheim Medical Faculty, Heidelberg University, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, Germany
| | - Dobromir Dobrev
- Institute of Pharmacology, West-German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Thomas Wieland
- Institute of Experimental and Clinical Pharmacology and Toxicology, Mannheim Medical Faculty, Heidelberg University, Mannheim, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, Germany
| |
Collapse
|
6
|
Lv Y, Xiao J, Liu J, Xing F. E2F8 is a Potential Therapeutic Target for Hepatocellular Carcinoma. J Cancer 2017; 8:1205-1213. [PMID: 28607595 PMCID: PMC5463435 DOI: 10.7150/jca.18255] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 01/11/2017] [Indexed: 12/15/2022] Open
Abstract
E2F transcriptional factors are widely expressed in a number of tissues and organs, possessing many regulatory functions related to cellular proliferation, differentiation, DNA repair, cell-cycle and cell apoptosis. E2F8 is a recently identified member of the E2F family with a duplicated DNA-binding domain feature discriminated from E2F1-6, controlling gene expression in a dimerization partner-independent manner. It is indispensable for angiogenesis, lymphangiogenesis and embryonic development. Although E2F8 and E2F7 perform complementary and overlapping functions in many cell metabolisms, E2F8, but not E2F7, overexpresses remarkably in hepatocellular carcinoma (HCC) to facilitate the HCC occurrence and development via activating a E2F1/ Cyclin D1 signaling pathway to regulate the G1- to S-phase transition of cell cycle progression or transcriptionally suppressing CDK1 to induce hepatocyte polyploidization. It also involves closely a variety of cellular physiological functions and pathological processes, which may bring a new breakthrough for the treatment of certain diseases, especially the HCC. Here, we summarize the latest progress of E2F8 on its relevant functions and mechanisms as well as potential application.
Collapse
Affiliation(s)
- Yi Lv
- Department of Immunobiology, Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou, China.,Key Laboratory of Functional Protein Research of Guangdong, Higher Education Institutes, Jinan University, Guangzhou, China
| | - Jia Xiao
- Department of Immunobiology, Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou, China
| | - Jing Liu
- Department of Stomatology, Jinan University, Guangzhou, China
| | - Feiyue Xing
- Department of Immunobiology, Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou, China.,Key Laboratory of Functional Protein Research of Guangdong, Higher Education Institutes, Jinan University, Guangzhou, China
| |
Collapse
|
7
|
Hetmann A, Wujak M, Kowalczyk S. Protein Transphosphorylation During the Mutual Interaction between Phytochrome A and a Nuclear Isoform of Nucleoside Diphosphate Kinase Is Regulated by Red Light. BIOCHEMISTRY (MOSCOW) 2017; 81:1153-1162. [PMID: 27908239 DOI: 10.1134/s0006297916100126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The nuclear isoform of nucleoside diphosphate kinase isoenzyme NDPK-In undergoes strong catalytic activation upon its interaction with the active form of phytochrome A (Pfr) in red light. The autophosphorylation or intermolecular transphosphorylation of NDPK-In leads to the formation of phosphoester bonds stable in acidic solution. The phosphate residue of the phosphamide bond in the active center of NDPK-In can also be transferred to serine and threonine residues localized in other proteins, including phytochrome A. Phytochrome A, similarly to NDPK-In, undergoes autophosphorylation on serine and threonine residues and can phosphorylate some potential substrate proteins. The physical interaction between phytochrome A in the Pfr form and NDPK-In results in a significant increase in the kinase activity of NDPK-In. The results presented in this work indicate that NDPK-In may function as a protein kinase regulated by light.
Collapse
Affiliation(s)
- A Hetmann
- Nicolaus Copernicus University, Faculty of Biology and Environment Protection, Department of Biochemistry, Toruń 87-100, Poland.
| | | | | |
Collapse
|
8
|
Qiu Y, Zhao D, Butenschön VM, Bauer AT, Schneider SW, Skolnik EY, Hammes HP, Wieland T, Feng Y. Nucleoside diphosphate kinase B deficiency causes a diabetes-like vascular pathology via up-regulation of endothelial angiopoietin-2 in the retina. Acta Diabetol 2016; 53:81-9. [PMID: 25900369 DOI: 10.1007/s00592-015-0752-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 03/30/2015] [Indexed: 11/25/2022]
Abstract
AIMS Nucleoside diphosphate kinase B (NDPKB) is capable of maintaining the cellular nucleotide triphosphate pools. It might therefore supply UTP for the formation of UDP-GlcNAc from glucose. As NDPKB contributes to vascular dysfunction, we speculate that NDPKB might play a role in microangiopathies, such as diabetic retinopathy (DR). Therefore, we investigated the impact of NDPKB on retinal vascular damage using NDPKB(-/-) mice during development of DR and its possible mechanisms. METHODS Pericyte loss and acellular capillary (AC) formation were assessed in streptozotocin-induced diabetic NDPKB(-/-) and wild-type (WT) mice. Expression of angiopoietin-2 (Ang2) and protein N-acetylglucosamine modification (GlcNAcylation) were assessed by western blot and/or immunofluorescence in the diabetic retinas as well as in endothelial cells depleted of NDPKB by siRNA and stimulated with high glucose. RESULTS Similar to diabetic WT retinas, non-diabetic NDPKB(-/-) retinas showed a significant decrease in pericyte coverage in comparison with non-diabetic WT retinas. Hyperglycemia further aggravates pericyte loss in diabetic NDPKB(-/-) retinas. AC formation was detected in the diabetic NDPKB(-/-) retinas. Similar to hyperglycemia, NDPKB deficiency induced Ang2 expression and protein GlcNAcylation that were not further altered in the diabetic retinas. In cultured endothelial cells, stimulation with high glucose and NDPKB depletion comparably increased Ang2 expression and protein GlcNAcylation. CONCLUSIONS Our data identify NDPKB as a protective factor in the retina, which controls Ang2 expression and the hexosamine pathway. NDPKB-deficient mice are a suitable model for studying mechanisms underlying diabetic retinal vascular damage.
Collapse
Affiliation(s)
- Yi Qiu
- Institute for Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Mybachstr. 14, 68169, Mannheim, Germany
| | - Di Zhao
- Institute for Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Mybachstr. 14, 68169, Mannheim, Germany
| | - Vicki-Marie Butenschön
- Institute for Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Mybachstr. 14, 68169, Mannheim, Germany
| | - Alexander T Bauer
- Division of Experimental Dermatology, Department of Dermatology, Venereology, and Allergology, Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Stefan W Schneider
- Division of Experimental Dermatology, Department of Dermatology, Venereology, and Allergology, Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Edward Y Skolnik
- Division of Nephrology, New York University Langone Medical Center, 560 1st Ave, New York, NY, 10016, USA
| | - Hans-Peter Hammes
- 5th Medical Clinic, Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Thomas Wieland
- Institute for Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Mybachstr. 14, 68169, Mannheim, Germany
| | - Yuxi Feng
- Institute for Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Mybachstr. 14, 68169, Mannheim, Germany.
| |
Collapse
|
9
|
Mishra S, Jakkala K, Srinivasan R, Arumugam M, Ranjeri R, Gupta P, Rajeswari H, Ajitkumar P. NDK Interacts with FtsZ and Converts GDP to GTP to Trigger FtsZ Polymerisation--A Novel Role for NDK. PLoS One 2015; 10:e0143677. [PMID: 26630542 PMCID: PMC4668074 DOI: 10.1371/journal.pone.0143677] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 11/09/2015] [Indexed: 11/19/2022] Open
Abstract
Introduction Nucleoside diphosphate kinase (NDK), conserved across bacteria to humans, synthesises NTP from NDP and ATP. The eukaryotic homologue, the NDPK, uses ATP to phosphorylate the tubulin-bound GDP to GTP for tubulin polymerisation. The bacterial cytokinetic protein FtsZ, which is the tubulin homologue, also uses GTP for polymerisation. Therefore, we examined whether NDK can interact with FtsZ to convert FtsZ-bound GDP and/or free GDP to GTP to trigger FtsZ polymerisation. Methods Recombinant and native NDK and FtsZ proteins of Mycobacterium smegmatis and Mycobacterium tuberculosis were used as the experimental samples. FtsZ polymersation was monitored using 90° light scattering and FtsZ polymer pelleting assays. The γ32P-GTP synthesised by NDK from GDP and γ32P-ATP was detected using thin layer chromatography and quantitated using phosphorimager. The FtsZ bound 32P-GTP was quantitated using phosphorimager, after UV-crosslinking, followed by SDS-PAGE. The NDK-FtsZ interaction was determined using Ni2+-NTA-pulldown assay and co-immunoprecipitation of the recombinant and native proteins in vitro and ex vivo, respectively. Results NDK triggered instantaneous polymerisation of GDP-precharged recombinant FtsZ in the presence of ATP, similar to the polymerisation of recombinant FtsZ (not GDP-precharged) upon the direct addition of GTP. Similarly, NDK triggered polymerisation of recombinant FtsZ (not GDP-precharged) in the presence of free GDP and ATP as well. Mutant NDK, partially deficient in GTP synthesis from ATP and GDP, triggered low level of polymerisation of MsFtsZ, but not of MtFtsZ. As characteristic of NDK’s NTP substrate non-specificity, it used CTP, TTP, and UTP also to convert GDP to GTP, to trigger FtsZ polymerisation. The NDK of one mycobacterial species could trigger the polymerisation of the FtsZ of another mycobacterial species. Both the recombinant and the native NDK and FtsZ showed interaction with each other in vitro and ex vivo, alluding to the possibility of direct phosphorylation of FtsZ-bound GDP by NDK. Conclusion Irrespective of the bacterial species, NDK interacts with FtsZ in vitro and ex vivo and, through the synthesis of GTP from FtsZ-bound GDP and/or free GDP, and ATP (CTP/TTP/UTP), triggers FtsZ polymerisation. The possible biological context of this novel activity of NDK is presented.
Collapse
Affiliation(s)
- Saurabh Mishra
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Kishor Jakkala
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Ramanujam Srinivasan
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Muthu Arumugam
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Raghavendra Ranjeri
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Prabuddha Gupta
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Haryadi Rajeswari
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Parthasarathi Ajitkumar
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
- * E-mail:
| |
Collapse
|
10
|
Zhou XB, Feng YX, Sun Q, Lukowski R, Qiu Y, Spiger K, Li Z, Ruth P, Korth M, Skolnik EY, Borggrefe M, Dobrev D, Wieland T. Nucleoside diphosphate kinase B-activated intermediate conductance potassium channels are critical for neointima formation in mouse carotid arteries. Arterioscler Thromb Vasc Biol 2015; 35:1852-61. [PMID: 26088577 DOI: 10.1161/atvbaha.115.305881] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 05/29/2015] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Vascular smooth muscle cells (VSMC) proliferation is a hallmark of atherosclerosis and vascular restenosis. The intermediate conductance Ca(2+)-activated K(+) (SK4) channel is required for pathological VSMC proliferation. In T lymphocytes, nucleoside diphosphate kinase B (NDPKB) has been implicated in SK4 channel activation. We thus investigated the role of NDPKB in the regulation of SK4 currents (ISK4) in proliferating VSMC and neointima formation. APPROACH AND RESULTS Function and expression of SK4 channels in VSMC from injured mouse carotid arteries were assessed by patch-clamping and real-time polymerase chain reaction. ISK4 was detectable in VSMC from injured but not from uninjured arteries correlating with the occurrence of the proliferative phenotype. Direct application of NDPKB to the membrane of inside-out patches increased ISK4, whereas NDPKB did not alter currents in VSMC obtained from injured vessels of SK4-deficient mice. The NDPKB-induced increase in ISK4 was prevented by protein histidine phosphatase 1, but not an inactive protein histidine phosphatase 1 mutant indicating that ISK4 is regulated via histidine phosphorylation in proliferating VSMC; moreover, genetic NDPKB ablation reduced ISK4 by 50% suggesting a constitutive activation of ISK4 in proliferating VSMC. In line, neointima formation after wire injury of the carotid artery was substantially reduced in mice deficient in SK4 channels or NDPKB. CONCLUSIONS NDPKB to SK4 signaling is required for neointima formation. Constitutive activation of SK4 by NDPKB in proliferating VSMC suggests that targeting this interaction via, for example, activation of protein histidine phosphatase 1 may provide clinically meaningful effects in vasculoproliferative diseases such as atherosclerosis and post angioplasty restenosis.
Collapse
Affiliation(s)
- Xiao-Bo Zhou
- From the 1st Medical Clinic (X.B.-Z., M.B.), Institute of Experimental and Clinical Pharmacology and Toxicology (Y.-X.F., Y.Q., K.S., T.W.), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Institute of Pharmacology, West German Heart and Vessel Centre, University Duisburg-Essen, Essen, Germany (Q.S., D.D.); Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany (R.L., P.R.); Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.K.); Department of Medicine (Z.L., E.Y.S.) and Department of Pharmacology (Z.L., E.Y.S.), Langone Medical Center, New York University; and DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Germany (M.B., T.W.)
| | - Yu-Xi Feng
- From the 1st Medical Clinic (X.B.-Z., M.B.), Institute of Experimental and Clinical Pharmacology and Toxicology (Y.-X.F., Y.Q., K.S., T.W.), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Institute of Pharmacology, West German Heart and Vessel Centre, University Duisburg-Essen, Essen, Germany (Q.S., D.D.); Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany (R.L., P.R.); Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.K.); Department of Medicine (Z.L., E.Y.S.) and Department of Pharmacology (Z.L., E.Y.S.), Langone Medical Center, New York University; and DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Germany (M.B., T.W.)
| | - Qiang Sun
- From the 1st Medical Clinic (X.B.-Z., M.B.), Institute of Experimental and Clinical Pharmacology and Toxicology (Y.-X.F., Y.Q., K.S., T.W.), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Institute of Pharmacology, West German Heart and Vessel Centre, University Duisburg-Essen, Essen, Germany (Q.S., D.D.); Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany (R.L., P.R.); Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.K.); Department of Medicine (Z.L., E.Y.S.) and Department of Pharmacology (Z.L., E.Y.S.), Langone Medical Center, New York University; and DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Germany (M.B., T.W.)
| | - Robert Lukowski
- From the 1st Medical Clinic (X.B.-Z., M.B.), Institute of Experimental and Clinical Pharmacology and Toxicology (Y.-X.F., Y.Q., K.S., T.W.), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Institute of Pharmacology, West German Heart and Vessel Centre, University Duisburg-Essen, Essen, Germany (Q.S., D.D.); Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany (R.L., P.R.); Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.K.); Department of Medicine (Z.L., E.Y.S.) and Department of Pharmacology (Z.L., E.Y.S.), Langone Medical Center, New York University; and DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Germany (M.B., T.W.)
| | - Yi Qiu
- From the 1st Medical Clinic (X.B.-Z., M.B.), Institute of Experimental and Clinical Pharmacology and Toxicology (Y.-X.F., Y.Q., K.S., T.W.), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Institute of Pharmacology, West German Heart and Vessel Centre, University Duisburg-Essen, Essen, Germany (Q.S., D.D.); Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany (R.L., P.R.); Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.K.); Department of Medicine (Z.L., E.Y.S.) and Department of Pharmacology (Z.L., E.Y.S.), Langone Medical Center, New York University; and DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Germany (M.B., T.W.)
| | - Katharina Spiger
- From the 1st Medical Clinic (X.B.-Z., M.B.), Institute of Experimental and Clinical Pharmacology and Toxicology (Y.-X.F., Y.Q., K.S., T.W.), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Institute of Pharmacology, West German Heart and Vessel Centre, University Duisburg-Essen, Essen, Germany (Q.S., D.D.); Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany (R.L., P.R.); Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.K.); Department of Medicine (Z.L., E.Y.S.) and Department of Pharmacology (Z.L., E.Y.S.), Langone Medical Center, New York University; and DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Germany (M.B., T.W.)
| | - Zhai Li
- From the 1st Medical Clinic (X.B.-Z., M.B.), Institute of Experimental and Clinical Pharmacology and Toxicology (Y.-X.F., Y.Q., K.S., T.W.), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Institute of Pharmacology, West German Heart and Vessel Centre, University Duisburg-Essen, Essen, Germany (Q.S., D.D.); Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany (R.L., P.R.); Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.K.); Department of Medicine (Z.L., E.Y.S.) and Department of Pharmacology (Z.L., E.Y.S.), Langone Medical Center, New York University; and DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Germany (M.B., T.W.)
| | - Peter Ruth
- From the 1st Medical Clinic (X.B.-Z., M.B.), Institute of Experimental and Clinical Pharmacology and Toxicology (Y.-X.F., Y.Q., K.S., T.W.), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Institute of Pharmacology, West German Heart and Vessel Centre, University Duisburg-Essen, Essen, Germany (Q.S., D.D.); Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany (R.L., P.R.); Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.K.); Department of Medicine (Z.L., E.Y.S.) and Department of Pharmacology (Z.L., E.Y.S.), Langone Medical Center, New York University; and DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Germany (M.B., T.W.)
| | - Michael Korth
- From the 1st Medical Clinic (X.B.-Z., M.B.), Institute of Experimental and Clinical Pharmacology and Toxicology (Y.-X.F., Y.Q., K.S., T.W.), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Institute of Pharmacology, West German Heart and Vessel Centre, University Duisburg-Essen, Essen, Germany (Q.S., D.D.); Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany (R.L., P.R.); Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.K.); Department of Medicine (Z.L., E.Y.S.) and Department of Pharmacology (Z.L., E.Y.S.), Langone Medical Center, New York University; and DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Germany (M.B., T.W.)
| | - Edward Y Skolnik
- From the 1st Medical Clinic (X.B.-Z., M.B.), Institute of Experimental and Clinical Pharmacology and Toxicology (Y.-X.F., Y.Q., K.S., T.W.), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Institute of Pharmacology, West German Heart and Vessel Centre, University Duisburg-Essen, Essen, Germany (Q.S., D.D.); Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany (R.L., P.R.); Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.K.); Department of Medicine (Z.L., E.Y.S.) and Department of Pharmacology (Z.L., E.Y.S.), Langone Medical Center, New York University; and DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Germany (M.B., T.W.)
| | - Martin Borggrefe
- From the 1st Medical Clinic (X.B.-Z., M.B.), Institute of Experimental and Clinical Pharmacology and Toxicology (Y.-X.F., Y.Q., K.S., T.W.), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Institute of Pharmacology, West German Heart and Vessel Centre, University Duisburg-Essen, Essen, Germany (Q.S., D.D.); Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany (R.L., P.R.); Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.K.); Department of Medicine (Z.L., E.Y.S.) and Department of Pharmacology (Z.L., E.Y.S.), Langone Medical Center, New York University; and DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Germany (M.B., T.W.)
| | - Dobromir Dobrev
- From the 1st Medical Clinic (X.B.-Z., M.B.), Institute of Experimental and Clinical Pharmacology and Toxicology (Y.-X.F., Y.Q., K.S., T.W.), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Institute of Pharmacology, West German Heart and Vessel Centre, University Duisburg-Essen, Essen, Germany (Q.S., D.D.); Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany (R.L., P.R.); Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.K.); Department of Medicine (Z.L., E.Y.S.) and Department of Pharmacology (Z.L., E.Y.S.), Langone Medical Center, New York University; and DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Germany (M.B., T.W.)
| | - Thomas Wieland
- From the 1st Medical Clinic (X.B.-Z., M.B.), Institute of Experimental and Clinical Pharmacology and Toxicology (Y.-X.F., Y.Q., K.S., T.W.), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Institute of Pharmacology, West German Heart and Vessel Centre, University Duisburg-Essen, Essen, Germany (Q.S., D.D.); Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany (R.L., P.R.); Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (M.K.); Department of Medicine (Z.L., E.Y.S.) and Department of Pharmacology (Z.L., E.Y.S.), Langone Medical Center, New York University; and DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg-Mannheim, Germany (M.B., T.W.).
| |
Collapse
|
11
|
Zhang P, Kofron CM, Mende U. Heterotrimeric G protein-mediated signaling and its non-canonical regulation in the heart. Life Sci 2015; 129:35-41. [PMID: 25818188 PMCID: PMC4415990 DOI: 10.1016/j.lfs.2015.02.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Revised: 01/31/2015] [Accepted: 02/11/2015] [Indexed: 11/20/2022]
Abstract
Heterotrimeric guanine nucleotide-binding proteins (G proteins) regulate a multitude of signaling pathways in mammalian cells by transducing signals from G protein-coupled receptors (GPCRs) to effectors, which in turn regulate cellular function. In the myocardium, G protein signaling occurs in all cardiac cell types and is centrally involved in the regulation of heart rate, pump function, and vascular tone and in the response to hemodynamic stress and injury. Perturbations in G protein-mediated signaling are well known to contribute to cardiac hypertrophy, failure, and arrhythmias. Most of the currently used drugs for cardiac and other diseases target GPCR signaling. In the canonical G protein signaling paradigm, G proteins that are located at the cytoplasmic surface of the plasma membrane become activated after an agonist-induced conformational change of GPCRs, which then allows GTP-bound Gα and free Gβγ subunits to activate or inhibit effector proteins. Research over the past two decades has markedly broadened the original paradigm with a GPCR-G protein-effector at the cell surface at its core by revealing novel binding partners and additional subcellular localizations for heterotrimeric G proteins that facilitate many previously unrecognized functional effects. In this review, we focus on non-canonical and epigenetic-related mechanisms that regulate heterotrimeric G protein expression, activation, and localization and discuss functional consequences using cardiac examples where possible. Mechanisms reviewed involve microRNAs, histone deacetylases, chaperones, alternative modes of G protein activation, and posttranslational modifications. Some of these newly characterized mechanisms may be further developed into novel strategies for the treatment of cardiac disease and beyond.
Collapse
Affiliation(s)
- Peng Zhang
- Cardiovascular Research Center, Cardiology Division, Rhode Island Hospital, Providence, RI, USA; Alpert Medical School of Brown University, Providence, RI, USA
| | - Celinda M Kofron
- Cardiovascular Research Center, Cardiology Division, Rhode Island Hospital, Providence, RI, USA; Alpert Medical School of Brown University, Providence, RI, USA
| | - Ulrike Mende
- Cardiovascular Research Center, Cardiology Division, Rhode Island Hospital, Providence, RI, USA; Alpert Medical School of Brown University, Providence, RI, USA.
| |
Collapse
|
12
|
Tong Y, Yung LY, Wong YH. Metastasis suppressors Nm23H1 and Nm23H2 differentially regulate neoplastic transformation and tumorigenesis. Cancer Lett 2015; 361:207-17. [PMID: 25748386 DOI: 10.1016/j.canlet.2015.02.050] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 02/25/2015] [Accepted: 02/26/2015] [Indexed: 11/30/2022]
Abstract
Nm23H1 and H2 are prototypical metastasis suppressors with diverse functions, but recent studies suggest that they may also regulate tumorigenesis. Here, we employed both cellular and in vivo assays to examine the effect of Nm23H1 and H2 on tumorigenesis induced by oncogenic Ras and/or p53 deficiency. Co-expression of Nm23H1 but not H2 in NIH3T3 cells effectively suppressed neoplastic transformation and tumorigenesis induced by the oncogenic H-Ras G12V mutant. Overexpression of Nm23H1 but not H2 also inhibited tumorigenesis by human cervical cancer HeLa cells with p53 deficiency. However, in human non-small-cell lung carcinoma H1299 cells harboring N-Ras Q61K oncogenic mutation and p53 deletion, overexpression of Nm23H1 did not affect tumorigenesis in nude mice assays, while overexpression of Nm23H2 enhanced tumor growth with elevated expression of the c-Myc proto-oncogene. Collectively, these results suggest that Nm23H1 and H2 have differential abilities to modulate tumorigenesis.
Collapse
Affiliation(s)
- Yao Tong
- Division of Life Sciences, Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Lisa Y Yung
- Division of Life Sciences, Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Yung H Wong
- Division of Life Sciences, Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong; State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong.
| |
Collapse
|
13
|
Kar A, Chowdhury S. Inhibition of telomerase activity by NME2: impact on metastasis suppression? Naunyn Schmiedebergs Arch Pharmacol 2014; 388:235-41. [PMID: 25547372 PMCID: PMC4469096 DOI: 10.1007/s00210-014-1077-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 11/25/2014] [Indexed: 12/25/2022]
Abstract
Though anti-metastatic function of non-metastatic 2 (NME2) has been implicated in multiple cancers, mechanisms of metastases control by NME2 are not clearly understood. Recent observations indicating the involvement of telomerase, the ribonucleoprotein required for telomere synthesis, in metastatic outcome are interesting. Notably, though the role of telomerase dysfunction in tumorigenesis is relatively well studied, involvement in metastasis progression is poorly understood. Recent findings demonstrate NME2 presence at telomere ends, association with telomerase, and NME2’s role in inhibition of telomerase activity in cancer cells. These present a novel opportunity to investigate mechanisms underlying NME2-mediated metastasis suppression.
Collapse
Affiliation(s)
- Anirban Kar
- Proteomics and Structural Biology Unit, CSIR-Institute of Genomics and Integrative Biology, Mathura Road, New Delhi, 110025, DELHI, India
| | | |
Collapse
|
14
|
Li Y, Tong Y, Wong YH. Regulatory functions of Nm23-H2 in tumorigenesis: insights from biochemical to clinical perspectives. Naunyn Schmiedebergs Arch Pharmacol 2014; 388:243-56. [PMID: 25413836 DOI: 10.1007/s00210-014-1066-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 11/07/2014] [Indexed: 12/12/2022]
Abstract
Substantial effort has been directed at elucidating the functions of the products of the Nm23 tumor metastasis suppressor genes over the past two decades, with the ultimate goal of exploring their translational potentials in changing cancer patients' outcomes. Much attention has been focused on the better-known Nm23-H1, but despite having high sequence similarity, Nm23-H2 functions differently in many aspects. Besides acting as a metastasis suppressor, compelling data suggest that Nm23-H2 may modulate various tumor-associated biological events to enhance tumorigenesis in human solid tumors and hematological malignancies. Linkage to tumorigenesis may occur through the ability of Nm23-H2 to regulate transcription, cell proliferation, apoptosis, differentiation, and telomerase activity. In this review, we examine the linkages of Nm23-H2 to tumorigenesis in terms of its biochemical and structural properties and discuss its potential role in various tumor-associated events.
Collapse
Affiliation(s)
- Yuanjun Li
- Division of Life Science and the Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | | | | |
Collapse
|
15
|
Feng Y, Gross S, Wolf NM, Butenschön VM, Qiu Y, Devraj K, Liebner S, Kroll J, Skolnik EY, Hammes HP, Wieland T. Nucleoside diphosphate kinase B regulates angiogenesis through modulation of vascular endothelial growth factor receptor type 2 and endothelial adherens junction proteins. Arterioscler Thromb Vasc Biol 2014; 34:2292-300. [PMID: 25147336 DOI: 10.1161/atvbaha.114.304239] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Nucleoside diphosphate kinase B (NDPKB) participates in the activation of heterotrimeric and monomeric G proteins, which are pivotal mediators in angiogenic signaling. The role of NDPKB in angiogenesis has to date not been defined. Therefore, we analyzed the contribution of NDPKB to angiogenesis and its underlying mechanisms in well-characterized in vivo and in vitro models. APPROACH AND RESULTS Zebrafish embryos were depleted of NDPKB by morpholino-mediated knockdown. These larvae displayed severe malformations specifically in vessels formed by angiogenesis. NDPKB-deficient (NDPKB(-/-)) mice were subjected to oxygen-induced retinopathy. In this model, the number of preretinal neovascularizations in NDPKB(-/-) mice was strongly reduced in comparison with wild-type littermates. In accordance, a delayed blood flow recovery was detected in the NDPKB(-/-) mice after hindlimb ligation. In in vitro studies, a small interfering RNA-mediated knockdown of NDPKB was performed in human umbilical endothelial cells. NDPKB depletion impaired vascular endothelial growth factor (VEGF)-induced sprouting and hampered the VEGF-induced spatial redistributions of the VEGF receptor type 2 and VE-cadherin at the plasma membrane. Concomitantly, NDPKB depletion increased the permeability of the human umbilical endothelial cell monolayer. CONCLUSIONS This is the first report to show that NDPKB is required for VEGF-induced angiogenesis and contributes to the correct localization of VEGF receptor type 2 and VE-cadherin at the endothelial adherens junctions. Therefore, our data identify NDPKB as a novel molecular target to modulate VEGF-dependent angiogenesis.
Collapse
Affiliation(s)
- Yuxi Feng
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (Y.F., S.G., N.M.W., V.M.B., Y.Q., T.W.), Department of Vascular Biology and Tumor Angiogenesis (J.K.), and the Fifth Medical Clinic (H.-P.H.), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Institute of Neurology (Edinger-Institute), Goethe University, Frankfurt, Germany (K.D., S.L.); and Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.)
| | - Shalini Gross
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (Y.F., S.G., N.M.W., V.M.B., Y.Q., T.W.), Department of Vascular Biology and Tumor Angiogenesis (J.K.), and the Fifth Medical Clinic (H.-P.H.), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Institute of Neurology (Edinger-Institute), Goethe University, Frankfurt, Germany (K.D., S.L.); and Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.)
| | - Nadine M Wolf
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (Y.F., S.G., N.M.W., V.M.B., Y.Q., T.W.), Department of Vascular Biology and Tumor Angiogenesis (J.K.), and the Fifth Medical Clinic (H.-P.H.), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Institute of Neurology (Edinger-Institute), Goethe University, Frankfurt, Germany (K.D., S.L.); and Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.)
| | - Vicki M Butenschön
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (Y.F., S.G., N.M.W., V.M.B., Y.Q., T.W.), Department of Vascular Biology and Tumor Angiogenesis (J.K.), and the Fifth Medical Clinic (H.-P.H.), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Institute of Neurology (Edinger-Institute), Goethe University, Frankfurt, Germany (K.D., S.L.); and Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.)
| | - Yi Qiu
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (Y.F., S.G., N.M.W., V.M.B., Y.Q., T.W.), Department of Vascular Biology and Tumor Angiogenesis (J.K.), and the Fifth Medical Clinic (H.-P.H.), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Institute of Neurology (Edinger-Institute), Goethe University, Frankfurt, Germany (K.D., S.L.); and Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.)
| | - Kavi Devraj
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (Y.F., S.G., N.M.W., V.M.B., Y.Q., T.W.), Department of Vascular Biology and Tumor Angiogenesis (J.K.), and the Fifth Medical Clinic (H.-P.H.), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Institute of Neurology (Edinger-Institute), Goethe University, Frankfurt, Germany (K.D., S.L.); and Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.)
| | - Stefan Liebner
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (Y.F., S.G., N.M.W., V.M.B., Y.Q., T.W.), Department of Vascular Biology and Tumor Angiogenesis (J.K.), and the Fifth Medical Clinic (H.-P.H.), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Institute of Neurology (Edinger-Institute), Goethe University, Frankfurt, Germany (K.D., S.L.); and Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.)
| | - Jens Kroll
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (Y.F., S.G., N.M.W., V.M.B., Y.Q., T.W.), Department of Vascular Biology and Tumor Angiogenesis (J.K.), and the Fifth Medical Clinic (H.-P.H.), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Institute of Neurology (Edinger-Institute), Goethe University, Frankfurt, Germany (K.D., S.L.); and Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.)
| | - Edward Y Skolnik
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (Y.F., S.G., N.M.W., V.M.B., Y.Q., T.W.), Department of Vascular Biology and Tumor Angiogenesis (J.K.), and the Fifth Medical Clinic (H.-P.H.), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Institute of Neurology (Edinger-Institute), Goethe University, Frankfurt, Germany (K.D., S.L.); and Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.)
| | - Hans-Peter Hammes
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (Y.F., S.G., N.M.W., V.M.B., Y.Q., T.W.), Department of Vascular Biology and Tumor Angiogenesis (J.K.), and the Fifth Medical Clinic (H.-P.H.), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Institute of Neurology (Edinger-Institute), Goethe University, Frankfurt, Germany (K.D., S.L.); and Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.)
| | - Thomas Wieland
- From the Institute of Experimental and Clinical Pharmacology and Toxicology (Y.F., S.G., N.M.W., V.M.B., Y.Q., T.W.), Department of Vascular Biology and Tumor Angiogenesis (J.K.), and the Fifth Medical Clinic (H.-P.H.), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Institute of Neurology (Edinger-Institute), Goethe University, Frankfurt, Germany (K.D., S.L.); and Division of Nephrology, New York University Langone Medical Center, New York (E.Y.S.).
| |
Collapse
|
16
|
Constitutive Activity of the Acetylcholine-Activated Potassium Current IK,ACh in Cardiomyocytes. ADVANCES IN PHARMACOLOGY 2014; 70:393-409. [DOI: 10.1016/b978-0-12-417197-8.00013-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
17
|
Marino N, Marshall JC, Collins JW, Zhou M, Qian Y, Veenstra T, Steeg PS. Nm23-h1 binds to gelsolin and inactivates its actin-severing capacity to promote tumor cell motility and metastasis. Cancer Res 2013; 73:5949-62. [PMID: 23940300 PMCID: PMC3825031 DOI: 10.1158/0008-5472.can-13-0368] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Nm23-H1 has been identified as a metastasis suppressor gene, but its protein interactions have yet to be understood with any mechanistic clarity. In this study, we evaluated the proteomic spectrum of interactions made by Nm23-H1 in 4T1 murine breast cancer cells derived from tissue culture, primary mammary tumors, and pulmonary metastases. By this approach, we identified the actin-severing protein Gelsolin as binding partner for Nm23-H1, verifying their interaction by coimmunoprecipitation in 4T1 cells as well as in human MCF7, MDA-MB-231T, and MDA-MB-435 breast cancer cells. In Gelsolin-transfected cells, coexpression of Nm23-H1 abrogated the actin-severing activity of Gelsolin. Conversely, actin severing by Gelsolin was abrogated by RNA interference-mediated silencing of endogenous Nm23-H1. Tumor cell motility was negatively affected in parallel with Gelsolin activity, suggesting that Nm23-H1 binding inactivated the actin-depolymerizing function of Gelsolin to inhibit cell motility. Using indirect immunoflourescence to monitor complexes formed by Gelsolin and Nm23-H1 in living cells, we observed their colocalization in a perinuclear cytoplasmic compartment that was associated with the presence of disrupted actin stress fibers. In vivo analyses revealed that Gelsolin overexpression increased the metastasis of orthotopically implanted 4T1 or tail vein-injected MDA-MB-231T cells (P = 0.001 and 0.04, respectively), along with the proportion of mice with diffuse liver metastases, an effect ablated by coexpression of Nm23-H1. We observed no variation in proliferation among lung metastases. Our findings suggest a new actin-based mechanism that can suppress tumor metastasis.
Collapse
MESH Headings
- Actins/metabolism
- Animals
- Apoptosis
- Blotting, Western
- Cell Line, Tumor
- Cell Movement
- Cell Proliferation
- Female
- Fluorescent Antibody Technique
- Gelsolin/antagonists & inhibitors
- Gelsolin/genetics
- Gelsolin/metabolism
- Gene Expression Regulation, Neoplastic
- Humans
- Immunoenzyme Techniques
- Immunoprecipitation
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/secondary
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/secondary
- Mammary Neoplasms, Animal/genetics
- Mammary Neoplasms, Animal/metabolism
- Mammary Neoplasms, Animal/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- NM23 Nucleoside Diphosphate Kinases/antagonists & inhibitors
- NM23 Nucleoside Diphosphate Kinases/genetics
- NM23 Nucleoside Diphosphate Kinases/metabolism
- RNA, Small Interfering/genetics
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
Collapse
Affiliation(s)
- Natascia Marino
- Women’s Cancers Section, Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda MD 20892
| | - Jean-Claude Marshall
- Women’s Cancers Section, Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda MD 20892
| | - Joshua W. Collins
- Women’s Cancers Section, Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda MD 20892
| | - Ming Zhou
- Laboratory of Proteomics and Analytical Technologies, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Yongzhen Qian
- Women’s Cancers Section, Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda MD 20892
| | - Timothy Veenstra
- Laboratory of Proteomics and Analytical Technologies, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Patricia S. Steeg
- Women’s Cancers Section, Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda MD 20892
| |
Collapse
|
18
|
Marino N, Marshall JC, Collins JW, Zhou M, Qian Y, Veenstra T, Steeg PS. Nm23-h1 binds to gelsolin and inactivates its actin-severing capacity to promote tumor cell motility and metastasis. Cancer Res 2013. [PMID: 23940300 DOI: 10.1158/0008-5472.can-13-0368-009-0109-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nm23-H1 has been identified as a metastasis suppressor gene, but its protein interactions have yet to be understood with any mechanistic clarity. In this study, we evaluated the proteomic spectrum of interactions made by Nm23-H1 in 4T1 murine breast cancer cells derived from tissue culture, primary mammary tumors, and pulmonary metastases. By this approach, we identified the actin-severing protein Gelsolin as binding partner for Nm23-H1, verifying their interaction by coimmunoprecipitation in 4T1 cells as well as in human MCF7, MDA-MB-231T, and MDA-MB-435 breast cancer cells. In Gelsolin-transfected cells, coexpression of Nm23-H1 abrogated the actin-severing activity of Gelsolin. Conversely, actin severing by Gelsolin was abrogated by RNA interference-mediated silencing of endogenous Nm23-H1. Tumor cell motility was negatively affected in parallel with Gelsolin activity, suggesting that Nm23-H1 binding inactivated the actin-depolymerizing function of Gelsolin to inhibit cell motility. Using indirect immunoflourescence to monitor complexes formed by Gelsolin and Nm23-H1 in living cells, we observed their colocalization in a perinuclear cytoplasmic compartment that was associated with the presence of disrupted actin stress fibers. In vivo analyses revealed that Gelsolin overexpression increased the metastasis of orthotopically implanted 4T1 or tail vein-injected MDA-MB-231T cells (P = 0.001 and 0.04, respectively), along with the proportion of mice with diffuse liver metastases, an effect ablated by coexpression of Nm23-H1. We observed no variation in proliferation among lung metastases. Our findings suggest a new actin-based mechanism that can suppress tumor metastasis.
Collapse
MESH Headings
- Actins/metabolism
- Animals
- Apoptosis
- Blotting, Western
- Cell Line, Tumor
- Cell Movement
- Cell Proliferation
- Female
- Fluorescent Antibody Technique
- Gelsolin/antagonists & inhibitors
- Gelsolin/genetics
- Gelsolin/metabolism
- Gene Expression Regulation, Neoplastic
- Humans
- Immunoenzyme Techniques
- Immunoprecipitation
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/secondary
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/secondary
- Mammary Neoplasms, Animal/genetics
- Mammary Neoplasms, Animal/metabolism
- Mammary Neoplasms, Animal/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- NM23 Nucleoside Diphosphate Kinases/antagonists & inhibitors
- NM23 Nucleoside Diphosphate Kinases/genetics
- NM23 Nucleoside Diphosphate Kinases/metabolism
- RNA, Small Interfering/genetics
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
Collapse
Affiliation(s)
- Natascia Marino
- Authors' Affiliations: Women's Cancers Section, Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda; and Laboratory of Proteomics and Analytical Technologies, Science Applications International Corporation-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | | | | | | | | | | | | |
Collapse
|
19
|
RGS19 inhibits Ras signaling through Nm23H1/2-mediated phosphorylation of the kinase suppressor of Ras. Cell Signal 2013; 25:1064-74. [PMID: 23416464 DOI: 10.1016/j.cellsig.2013.02.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 02/09/2013] [Indexed: 12/31/2022]
Abstract
Besides serving as signal terminators for G protein pathways, several regulators of G protein signaling (RGS) can also modulate cell proliferation. RGS19 has previously been shown to enhance Akt signaling despite impaired Ras signaling. The present study examines the mechanism by which RGS19 inhibits Ras signaling. In HEK293 cells stably expressing RGS19, serum-induced Ras activation and phosphorylations of Raf/MEK/ERK were significantly inhibited, while cells expressing RGS2, 4, 7, 8, 10, or 20 did not exhibit this inhibitory phenotype. Conversely, siRNA-mediated knockdown of RGS19 enabled partial recovery of serum-induced ERK phosphorylation. Interestingly, two isoforms of the tumor metastasis suppressor Nm23 (H1 and H2) were upregulated in 293/RGS19 cells. As a nucleoside diphosphate kinase, Nm23H1 can phosphorylate the kinase suppressor of Ras (KSR). Elevated levels of phosphorylated KSR were indeed detected in the nuclear fractions of 293/RGS19 cells. Co-immunoprecipitation assays revealed that Nm23H1/2 can form complexes with RGS19, Ras, or KSR. siRNA-mediated knockdown of Nm23H1/2 allowed 293/RGS19 cells to partially recover their ERK responses to serum treatment, while overexpression of Nm23H1/2 in HEK293 cells suppressed the serum-induced ERK response. This study demonstrates that expression of RGS19 can suppress Ras-mediated signaling via upregulation of Nm23.
Collapse
|
20
|
Liu CY, Tao S, Xue JY, Zhang H, Xue WT, Chen FS. Identification and purification of a novel fish allergen from largemouth bass (Micropterus salmoides). FOOD AGR IMMUNOL 2012. [DOI: 10.1080/09540105.2012.745122] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
21
|
Arai S, Yonezawa Y, Okazaki N, Matsumoto F, Tamada T, Tokunaga H, Ishibashi M, Blaber M, Tokunaga M, Kuroki R. A structural mechanism for dimeric to tetrameric oligomer conversion in Halomonas sp. nucleoside diphosphate kinase. Protein Sci 2012; 21:498-510. [PMID: 22275000 DOI: 10.1002/pro.2032] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Accepted: 01/19/2012] [Indexed: 11/11/2022]
Abstract
Nucleoside diphosphate kinase (NDK) is known to form homotetramers or homohexamers. To clarify the oligomer state of NDK from moderately halophilic Halomonas sp. 593 (HaNDK), the oligomeric state of HaNDK was characterized by light scattering followed by X-ray crystallography. The molecular weight of HaNDK is 33,660, and the X-ray crystal structure determination to 2.3 and 2.7 Å resolution showed a dimer form which was confirmed in the different space groups of R3 and C2 with an independent packing arrangement. This is the first structural evidence that HaNDK forms a dimeric assembly. Moreover, the inferred molecular mass of a mutant HaNDK (E134A) indicated 62.1-65.3 kDa, and the oligomerization state was investigated by X-ray crystallography to 2.3 and 2.5 Å resolution with space groups of P2(1) and C2. The assembly form of the E134A mutant HaNDK was identified as a Type I tetramer as found in Myxococcus NDK. The structural comparison between the wild-type and E134A mutant HaNDKs suggests that the change from dimer to tetramer is due to the removal of negative charge repulsion caused by the E134 in the wild-type HaNDK. The higher ordered association of proteins usually contributes to an increase in thermal stability and substrate affinity. The change in the assembly form by a minimum mutation may be an effective way for NDK to acquire molecular characteristics suited to various circumstances.
Collapse
Affiliation(s)
- Shigeki Arai
- Molecular Structural Biology Group, Quantum Beam Science Directorate, Japan Atomic Energy Agency, 2-4 Shirakata-Shirane, Tokai, Ibaraki 319-1195, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Thakur RK, Yadav VK, Kumar P, Chowdhury S. Mechanisms of non-metastatic 2 (NME2)-mediated control of metastasis across tumor types. Naunyn Schmiedebergs Arch Pharmacol 2011; 384:397-406. [PMID: 21556888 DOI: 10.1007/s00210-011-0631-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2011] [Accepted: 03/25/2011] [Indexed: 12/18/2022]
Abstract
Non-metastatic 23 [NM23/nucleoside diphosphate kinases (NDPK)] genes are the first discovered metastasis suppressor genes. More than two decades of research has demonstrated their roles in a variety of biological processes with NME1 and NME2 being most studied in the context of metastasis suppression. Although NME1 and NME2 share >85% homology at amino acid level, they show redundant as well as unique molecular functions. Phenotypic analyses of knockout (KO) mice for NM23 members (NDPK-A, B) and compound KO (A as well as B) showed requirement of both proteins in hematopoiesis suggesting shared functions in development disease. Several reviews have discussed NME1, however the role of NME2 appears to be relatively less understood in the context of metastasis suppression. Here, we focus on NME2 and by meta-analysis of gene expression from multiple tumor types, and survey of in vivo and vitro studies, suggest the possibility that NME2 may be one of the key factors in metastasis. This along with the relevance of normal physiological functions of NME2 in the context of metastasis is discussed. We further examined the genetic and epigenetic features of NME2 and NME1 gene promoters and found aspects of transcription control that could be unique to NME2/NME1. Findings on signaling pathways and small molecules which regulate the expression of NME2 that could be therapeutically important are also discussed.
Collapse
Affiliation(s)
- Ram Krishna Thakur
- Proteomics and Structural Biology Unit, Institute of Genomics and Integrative Biology, CSIR, Mall Road, Delhi, 110 007, India
| | | | | | | |
Collapse
|
23
|
Nucleoside diphosphate kinase B is required for the formation of heterotrimeric G protein containing caveolae. Naunyn Schmiedebergs Arch Pharmacol 2011; 384:461-72. [PMID: 21409430 DOI: 10.1007/s00210-011-0618-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2011] [Accepted: 02/25/2011] [Indexed: 01/12/2023]
Abstract
Caveolae are flask-shaped invaginations in the plasma membrane that serve to compartmentalize and organize signal transduction processes, including signals mediated by G protein-coupled receptors and heterotrimeric G proteins. Herein we report evidence for a close association of the nucleoside diphosphate kinase B (NDPK B) and caveolin proteins which is required for G protein scaffolding and caveolae formation. A concomitant loss of the proteins NDPK B, caveolin isoforms 1 (Cav1) and 3, and heterotrimeric G proteins occurred when one of these proteins was specifically depleted in zebrafish embryos. Co-immunoprecipitation of Cav1 with the G protein Gβ-subunit and NDPK B from zebrafish lysates corroborated the direct association of these proteins. Similarly, in embryonic fibroblasts from the respective knockout (KO) mice, the membrane content of the Cav1, Gβ, and NDPK B was found to be mutually dependent on one another. A redistribution of Cav1 and Gβ from the caveolae containing fractions of lower density to other membrane compartments with higher density could be detected by means of density gradient fractionation of membranes derived from NDPK A/B KO mouse embryonic fibroblasts (MEFs) and after shRNA-mediated NDPK B knockdown in H10 cardiomyocytes. This redistribution could be visualized by confocal microscopy analysis showing a decrease in the plasma membrane bound Cav1 in NDPK A/B KO cells and vice versa and a decrease in the plasma membrane pool of NDPK B in Cav1 KO cells. Consequently, ultrastructural analysis revealed a reduction of surface caveolae in the NDPK A/B KO cells. To prove that the disturbed subcellular localization of Cav1 in NDPK A/B KO MEFs as well as NDPK B in Cav1 KO MEFs is a result of the loss of NDPK B and Cav1, respectively, we performed rescue experiments. The adenoviral re-expression of NDPK B in NDPK A/B KO MEFs rescued the protein content and the plasma membrane localization of Cav1. The expression of an EGFP-Cav1 fusion protein in Cav1-KO cells induced a restoration of NDPK B expression levels and its appearance at the plasma membrane. We conclude from these findings that NDPK B, heterotrimeric G proteins, and caveolins are mutually dependent on each other for stabile localization and caveolae formation at the plasma membrane. The data point to a disturbed transport of caveolin/G protein/NDPK B complexes from intracellular membrane compartments if one of the components is missing.
Collapse
|
24
|
Wang QQ, Zhao X, Pu XP. Proteome analysis of the left ventricle in the vitamin D₃ and nicotine-induced rat vascular calcification model. J Proteomics 2011; 74:480-9. [PMID: 21237295 DOI: 10.1016/j.jprot.2010.12.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Revised: 12/13/2010] [Accepted: 12/28/2010] [Indexed: 11/17/2022]
Abstract
Vitamin D₃ and nicotine (VDN) serve as an animal model of arterial calcification. The vascular calcification induced by the VDN model is always accompanied by compensatory left ventricular (LV) hypertrophy and impaired cardiac performance. To determine the possible mechanisms that are responsible for the effects of VDN on the LV, a 2-DE based proteomics approach was used to evaluate the changes in protein expression of the left ventricle in VDN rats, to our knowledge, for the first time. We identified sixteen proteins that were markedly altered and involved in mitochondrial function, heat shock protein activity, myocyte cytoskeleton composition and enzyme activity for energy metabolism. We describe, for the first time, a novel pathway (NDPK) that is involved in LV hypertrophy and enzyme activities of three of the sixteen clinical identified proteins: lactate dehydrogenase (LDH), SOD [Mn] and GST.
Collapse
Affiliation(s)
- Qian-Qian Wang
- National Key Research laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100083, PR China
| | | | | |
Collapse
|
25
|
Völkers M, Weidenhammer C, Herzog N, Qiu G, Spaich K, Wegner FV, Peppel K, Müller OJ, Schinkel S, Rabinowitz JE, Hippe HJ, Brinks H, Katus HA, Koch WJ, Eckhart AD, Friedrich O, Most P. The inotropic peptide βARKct improves βAR responsiveness in normal and failing cardiomyocytes through G(βγ)-mediated L-type calcium current disinhibition. Circ Res 2011; 108:27-39. [PMID: 21106943 PMCID: PMC4013502 DOI: 10.1161/circresaha.110.225201] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2009] [Accepted: 11/15/2010] [Indexed: 12/20/2022]
Abstract
RATIONALE The G(βγ)-sequestering peptide β-adrenergic receptor kinase (βARK)ct derived from the G-protein-coupled receptor kinase (GRK)2 carboxyl terminus has emerged as a promising target for gene-based heart failure therapy. Enhanced downstream cAMP signaling has been proposed as the underlying mechanism for increased β-adrenergic receptor (βAR) responsiveness. However, molecular targets mediating improved cardiac contractile performance by βARKct and its impact on G(βγ)-mediated signaling have yet to be fully elucidated. OBJECTIVE We sought to identify G(βγ)-regulated targets and signaling mechanisms conveying βARKct-mediated enhanced βAR responsiveness in normal (NC) and failing (FC) adult rat ventricular cardiomyocytes. METHODS AND RESULTS Assessing viral-based βARKct gene delivery with electrophysiological techniques, analysis of contractile performance, subcellular Ca²(+) handling, and site-specific protein phosphorylation, we demonstrate that βARKct enhances the cardiac L-type Ca²(+) channel (LCC) current (I(Ca)) both in NCs and FCs on βAR stimulation. Mechanistically, βARKct augments I(Ca) by preventing enhanced inhibitory interaction between the α1-LCC subunit (Cav1.2α) and liberated G(βγ) subunits downstream of activated βARs. Despite improved βAR contractile responsiveness, βARKct neither increased nor restored cAMP-dependent protein kinase (PKA) and calmodulin-dependent kinase II signaling including unchanged protein kinase (PK)Cε, extracellular signal-regulated kinase (ERK)1/2, Akt, ERK5, and p38 activation both in NCs and FCs. Accordingly, although βARKct significantly increases I(Ca) and Ca²(+) transients, being susceptible to suppression by recombinant G(βγ) protein and use-dependent LCC blocker, βARKct-expressing cardiomyocytes exhibit equal basal and βAR-stimulated sarcoplasmic reticulum Ca²(+) load, spontaneous diastolic Ca²(+) leakage, and survival rates and were less susceptible to field-stimulated Ca²(+) waves compared with controls. CONCLUSION Our study identifies a G(βγ)-dependent signaling pathway attenuating cardiomyocyte I(Ca) on βAR as molecular target for the G(βγ)-sequestering peptide βARKct. Targeted interruption of this inhibitory signaling pathway by βARKct confers improved βAR contractile responsiveness through increased I(Ca) without enhancing regular or restoring abnormal cAMP-signaling. βARKct-mediated improvement of I(Ca) rendered cardiomyocytes neither susceptible to βAR-induced damage nor arrhythmogenic sarcoplasmic reticulum Ca²(+) leakage.
Collapse
Affiliation(s)
- Mirko Völkers
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Christian Weidenhammer
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Nicole Herzog
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Gang Qiu
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Kristin Spaich
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Frederic V Wegner
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Karsten Peppel
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Oliver J Müller
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Stefanie Schinkel
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Joseph E Rabinowitz
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Hans-Jorg Hippe
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Henriette Brinks
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Hugo A Katus
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Walter J Koch
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Andrea D Eckhart
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Oliver Friedrich
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| | - Patrick Most
- Center for Molecular and Translational Cardiology (M.V, C.W., N.H., K.S., P.M.), Department of Internal Medicine III (O.J.M, S.S., H.J.H., H.A.K.), Division of Cardiology, INF 350, University of Heidelberg, 69120 Heidelberg, Germany; Institute of Physiology and Pathophysiology (F.W., O.F.) Medical Biophysics, INF 326, University of Heidelberg, 69120 Heidelberg, Germany; George Zallie & Family Laboratory for Cardiovascular Gene Therapy (J.E.R., H.B., W.J.K.), Eugene Feiner Laboratory for Vascular Biology and Thrombosis (A.D.E.), Laboratory for Cardiac Stem Cell and Gene Therapy (G.Q., K.P., P.M.), Center for Translational Medicine, Thomas Jefferson University, 19107 Philadelphia, PA, USA
| |
Collapse
|
26
|
Dharmasiri S, Harrington HM, Dharmasiri N. Heat shock modulates phosphorylation status and activity of nucleoside diphosphate kinase in cultured sugarcane cells. PLANT CELL REPORTS 2010; 29:1305-14. [PMID: 20821213 DOI: 10.1007/s00299-010-0917-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Revised: 08/17/2010] [Accepted: 08/23/2010] [Indexed: 05/29/2023]
Abstract
Nucleoside diphosphate kinase (NDPK) is involved in the regeneration of nucleoside triphosphates (NTPs) through its phosphotransferase activity via an autophosphorylating histidine residue. Additionally, autophosphorylation of serine and/or threonine residues is documented for NDPKs from various organisms. However, the metabolic significance of serine/threonine phosphorylation has not been well characterized. In this study we report the cloning and characterization of NDPKI from cultured sugarcane (Saccharum officinarum L. line H50-7209) cells, and modulation of serine autophosphorylation of NDPK1 in response to heat-shock (HS). Heat-shock treatment at 40°C for 2 h resulted in a 40% reduction in labeled phosphoserine in NDPK1. This dephosphorylation was accompanied by an increase in NDPK enzyme activity. In contrast, NDPK1 in cultured tobacco (cv. W-38) cells did not show changes in autophosphorylation or increased enzyme activity in response to HS. The mRNA or protein level of NDPK1 did not increase in response to HS. Sugarcane cells sustain the constitutive protein synthesis in addition to heat-shock protein synthesis during HS, while constitutive protein synthesis is significantly reduced in tobacco cells during HS. Thus, HS modulation of NDPK1 activity and serine dephosphorylation in sugarcane cells may represent an important physiological role in maintaining cellular metabolic functions during heat stress.
Collapse
Affiliation(s)
- Sunethra Dharmasiri
- Department of Biology, Texas State University, 601, University Drive, San Marcos, USA.
| | | | | |
Collapse
|
27
|
Mehta A, Orchard S. Nucleoside diphosphate kinase (NDPK, NM23, AWD): recent regulatory advances in endocytosis, metastasis, psoriasis, insulin release, fetal erythroid lineage and heart failure; translational medicine exemplified. Mol Cell Biochem 2009; 329:3-15. [PMID: 19415463 PMCID: PMC2721137 DOI: 10.1007/s11010-009-0114-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2009] [Accepted: 04/02/2009] [Indexed: 12/17/2022]
Abstract
The guest editor (AM) provides his perspective on the most recent advances on nucleoside diphosphate kinase (NDPK, otherwise known as AWD or NM23) showcasing phospho-histidine biochemistry and its impact on diverse pathology when disordered. His co-author (SO) provides state-of-the-art analyses from the European institute of Bioinformatics in an appendix to support the most recent advances made by the NDPK community. Unfortunately, to those outside the field, NDPK is often dismissed as a tiny ‘ancient housekeeper’ protein found in marine sponges, social amoebae, worms, fruit flies, rodents and humans but the state-of-the-art papers overviewed here show that NDPK does not act simply in mindless rote, inter-converting cellular ‘energy currencies’. That two NDPK isoforms regulate fetal erythroid lineage is a developmental case in point. Seminal Cancer Research UK support is gratefully acknowledged that generated additional resources to enable the NDPK community to meet in Dundee in 2007 (www.dundee.ac.uk/mchs/ndpk; next meeting is planned: 2010/Mannheim-Heidelberg). The presented papers illustrate the point that when scientists are left alone ‘shut up in the narrow cell of their laboratory’ (as the philosopher Ortega once said, a sentiment echoed by Erwin Schrödinger), then progress will ultimately occur bridging the gap between specialization and translation for human benefit. To aid translation, this overview initially introduces the NDPK family to the non-specialist, who serendipitously finds these proteins in their biology. This is immediately followed by examples of the diverse biology generated by this self-aggregating group of multi-functional proteins and finally capped by an emerging idea explaining how this diversity might arise.
Collapse
Affiliation(s)
- Anil Mehta
- Division of Medical Sciences, Tayside Institute of Child Health, Ninewells Hospital Medical School, Dundee DD19SY, Scotland, UK.
| | | |
Collapse
|
28
|
Mitochondrial kinases and their molecular interaction with cardiolipin. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2009; 1788:2032-47. [PMID: 19409873 DOI: 10.1016/j.bbamem.2009.04.018] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2008] [Accepted: 04/24/2009] [Indexed: 11/22/2022]
Abstract
Mitochondrial isoforms of creatine kinase (MtCK) and nucleoside diphosphate kinase (NDPK-D) are not phylogenetically related but share functionally important properties. They both use mitochondrially generated ATP with the ultimate goal of maintaining proper nucleotide pools, are located in the intermembrane/cristae space, have symmetrical oligomeric structures, and show high affinity binding to anionic phospholipids, in particular cardiolipin. The structural basis and functional consequences of the cardiolipin interaction have been studied and are discussed in detail in this review. They mainly result in a functional interaction of MtCK and NDPK-D with inner membrane adenylate translocator, probably by forming proteolipid complexes. These interactions allow for privileged exchange of metabolites (channeling) that ultimately regulate mitochondrial respiration. Further functions of the MtCK/membrane interaction include formation of cardiolipin membrane patches, stabilization of mitochondria and a role in apoptotic signaling, as well as in case of both kinases, a role in facilitating lipid transfer between two membranes. Finally, disturbed cardiolipin interactions of MtCK, NDPK-D and other proteins like cytochrome c and truncated Bid are discussed more generally in the context of apoptosis and necrosis.
Collapse
|
29
|
Dupré DJ, Robitaille M, Rebois RV, Hébert TE. The role of Gbetagamma subunits in the organization, assembly, and function of GPCR signaling complexes. Annu Rev Pharmacol Toxicol 2009; 49:31-56. [PMID: 18834311 DOI: 10.1146/annurev-pharmtox-061008-103038] [Citation(s) in RCA: 212] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The role of Gbetagamma subunits in cellular signaling has become well established in the past 20 years. Not only do they regulate effectors once thought to be the sole targets of Galpha subunits, but it has become clear that they also have a unique set of binding partners and regulate signaling pathways that are not always localized to the plasma membrane. However, this may be only the beginning of the story. Gbetagamma subunits interact with G protein-coupled receptors, Galpha subunits, and several different effector molecules during assembly and trafficking of receptor-based signaling complexes and not simply in response to ligand stimulation at sites of receptor cellular activity. Gbetagamma assembly itself seems to be tightly regulated via the action of molecular chaperones and in turn may serve a similar role in the assembly of specific signaling complexes. We propose that specific Gbetagamma subunits have a broader role in controlling the architecture, assembly, and activity of cellular signaling pathways.
Collapse
Affiliation(s)
- Denis J Dupré
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada.
| | | | | | | |
Collapse
|
30
|
Abstract
Nucleoside diphosphate kinases (NDPK) are encoded by the NME genes, also called NM23. They catalyze the transfer of gamma-phosphate from nucleoside triphosphates to nucleoside diphosphates by a ping-pong mechanism involving the formation of a high energy phospho-histidine intermediate [1, 2]. Besides their known functions in the control of intracellular nucleotide homeostasis, they are involved in multiple physiological and pathological cellular processes such as differentiation, development, metastastic dissemination or cilia functions. Over the past 15 years, ten human genes have been discovered encoding partial, full length, and/or tandemly repeated Nm23/NDPK domains, with or without N-or C-terminal extensions and/or additional domains. These genes encode proteins exhibiting different functions at various tissular and subcellular localizations. Most of these genes appear late in evolution with the emergence of the vertebrate lineage. This review summarizes the present knowledge on these multitalented proteins.
Collapse
|
31
|
Boissan M, Dabernat S, Peuchant E, Schlattner U, Lascu I, Lacombe ML. The mammalian Nm23/NDPK family: from metastasis control to cilia movement. Mol Cell Biochem 2009; 329:51-62. [PMID: 19387795 DOI: 10.1007/s11010-009-0120-7] [Citation(s) in RCA: 155] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2009] [Accepted: 04/02/2009] [Indexed: 01/12/2023]
Abstract
Nucleoside diphosphate kinases (NDPK) are encoded by the NME genes, also called NM23. They catalyze the transfer of gamma-phosphate from nucleoside triphosphates to nucleoside diphosphates by a ping-pong mechanism involving the formation of a high energy phospho-histidine intermediate [1, 2]. Besides their known functions in the control of intracellular nucleotide homeostasis, they are involved in multiple physiological and pathological cellular processes such as differentiation, development, metastastic dissemination or cilia functions. Over the past 15 years, ten human genes have been discovered encoding partial, full length, and/or tandemly repeated Nm23/NDPK domains, with or without N-or C-terminal extensions and/or additional domains. These genes encode proteins exhibiting different functions at various tissular and subcellular localizations. Most of these genes appear late in evolution with the emergence of the vertebrate lineage. This review summarizes the present knowledge on these multitalented proteins.
Collapse
Affiliation(s)
- Mathieu Boissan
- INSERM UMRS_938, UMPC Université Paris 06, 75012 Paris, France
| | | | | | | | | | | |
Collapse
|
32
|
Koeck T, Willard B, Crabb JW, Kinter M, Stuehr DJ, Aulak KS. Glucose-mediated tyrosine nitration in adipocytes: targets and consequences. Free Radic Biol Med 2009; 46:884-92. [PMID: 19135148 PMCID: PMC2888280 DOI: 10.1016/j.freeradbiomed.2008.12.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2008] [Revised: 12/03/2008] [Accepted: 12/11/2008] [Indexed: 12/20/2022]
Abstract
Hyperglycemia, a key factor in insulin resistance and diabetic pathology, is associated with cellular oxidative stress that promotes oxidative protein modifications. We report that protein nitration is responsive to changes in glucose concentrations in 3T3-L1 adipocytes. Alterations in the extent of tyrosine nitration as well as the cellular nitroproteome profile correlated tightly with changing glucose concentrations. The target proteins we identified are involved in fatty acid binding, cell signaling, protein folding, energy metabolism, antioxidant capacity, and membrane permeability. The nitration of adipocyte fatty acid binding protein (FABP4) at Tyr19 decreases, similar to phosphorylation, the binding of palmitic acid to the fatty acid-free protein. This potentially alters intracellular fatty acid transport, nuclear translocation of FABP4, and agonism of PPAR gamma. Our results suggest that protein tyrosine nitration may be a factor in obesity, insulin resistance, and the pathogenesis of diabetes.
Collapse
Affiliation(s)
- Thomas Koeck
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44118, USA
- Corresponding authors. Fax: +1 216 444 8372. (T. Koeck), (K.S. Aulak)
| | - Belinda Willard
- Department of Cell Biology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - John W. Crabb
- Departments of Ophthalmic Research and Cell Biology, Cole Eye Institute and Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Mike Kinter
- Department of Cell Biology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Dennis J. Stuehr
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44118, USA
| | - Kulwant S. Aulak
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44118, USA
- Corresponding authors. Fax: +1 216 444 8372. (T. Koeck), (K.S. Aulak)
| |
Collapse
|
33
|
Mitchell KAP, Szabo G, de S Otero A. Direct binding of cytosolic NDP kinases to membrane lipids is regulated by nucleotides. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2008; 1793:469-76. [PMID: 19146889 DOI: 10.1016/j.bbamcr.2008.12.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2008] [Revised: 11/30/2008] [Accepted: 12/03/2008] [Indexed: 11/19/2022]
Abstract
In spite of their complete lack of any structural features that characterize membrane proteins, cytosolic nucleoside-diphosphate kinases (NDPKs) have been found repeatedly to associate with membranes. In some instances the recruitment of cytosolic NDPKs to membranes was attributed to interactions with peripheral or integral membrane proteins, but in many cases the mechanism underlying the association of NDPKs with membranes remained unknown. We show here that cytosolic NDPKs bind directly to membrane lipids in a dynamic process that is controlled by its substrates, nucleoside tri- and diphosphates, and can be fully reconstituted with chemically defined, protein-free phospholipids and recombinant NDPK, or with purified NDPK. Our results uncover a novel mechanism for the reversible targeting of soluble NDPKs to membranes, where they may act as a reservoir of high energy phosphate, supporting the operation of membrane-based processes that utilize nucleotides other than ATP, such as intracellular traffic and phospholipid biosynthesis.
Collapse
Affiliation(s)
- Kimberly A P Mitchell
- Department of Molecular Physiology and Biological Physics, University of Virginia Medical School, P.O. Box 800736, Charlottesville, VA 22908-0736, USA
| | | | | |
Collapse
|
34
|
Tokunaga H, Arakawa T, Tokunaga M. Engineering of halophilic enzymes: two acidic amino acid residues at the carboxy-terminal region confer halophilic characteristics to Halomonas and Pseudomonas nucleoside diphosphate kinases. Protein Sci 2008; 17:1603-10. [PMID: 18573868 DOI: 10.1110/ps.035725.108] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Nucleoside diphosphate kinase from Halomonas sp. 593 (HaNDK) exhibits halophilic characteristics. Residues 134 and 135 in the carboxy-terminal region of HaNDK are Glu-Glu, while those of its homologous counterpart of non-halophilic Pseudomonas NDK (PaNDK) are Ala-Ala. The double mutation, E134A-E135A, in HaNDK results in the loss of the halophilic characteristics, and, conversely, the double mutation of A134E-A135E in PaNDK confers halophilic characters to this enzyme, indicating that the charged state of these two residues that are located in the C-terminal region plays a critical role in determining halophilic characteristics. The importance of these two residues versus the net negative charges will be discussed in relation to the halophilicity of NDK.
Collapse
Affiliation(s)
- Hiroko Tokunaga
- Applied and Molecular Microbiology, Faculty of Agriculture, Kagoshima University, Kagoshima 890-0065, Japan
| | | | | |
Collapse
|
35
|
Mailloux RJ, Darwich R, Lemire J, Appanna V. The monitoring of nucleotide diphosphate kinase activity by blue native polyacrylamide gel electrophoresis. Electrophoresis 2008; 29:1484-9. [DOI: 10.1002/elps.200700697] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
36
|
Tokunaga H, Ishibashi M, Arisaka F, Arai S, Kuroki R, Arakawa T, Tokunaga M. Residue 134 determines the dimer-tetramer assembly of nucleoside diphosphate kinase from moderately halophilic bacteria. FEBS Lett 2008; 582:1049-54. [DOI: 10.1016/j.febslet.2008.02.054] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2008] [Revised: 02/22/2008] [Accepted: 02/23/2008] [Indexed: 12/01/2022]
|
37
|
Mehta A. The cystic fibrosis transmembrane recruiter the alter ego of CFTR as a multi-kinase anchor. Pflugers Arch 2007; 455:215-21. [PMID: 17805562 PMCID: PMC2629509 DOI: 10.1007/s00424-007-0290-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2006] [Revised: 04/27/2007] [Accepted: 05/11/2007] [Indexed: 11/03/2022]
Abstract
This review focuses on a newly discovered interaction between protein kinases involved in cellular energetics, a process that may be disturbed in cystic fibrosis for unknown reasons. I propose a new model where kinase-mediated cellular transmission of energy provides mechanistic insight to a latent role of the cystic fibrosis transmembrane conductance regulator (CFTR). I suggest that CFTR acts as a multi-kinase recruiter to the apical epithelial membrane. My group finds that, in the cytosol, two protein kinases involved in cell energy homeostasis, nucleoside diphosphate kinase (NDPK) and AMP-activated kinase (AMPK), bind one another. Preliminary data suggest that both can also bind CFTR (function unclear). The disrupted role of this CFTR-kinase complex as ‘membrane transmitter to the cell’ is proposed as an alternative paradigm to the conventional ion transport mediated and CFTR/chloride-centric view of cystic fibrosis pathogenesis. Chloride remains important, but instead, chloride-induced control of the phosphohistidine content of one kinase component (NDPK, via a multi-kinase complex that also includes a third kinase, CK2; formerly casein kinase 2). I suggest that this complex provides the necessary near-equilibrium conditions needed for efficient transmission of phosphate energy to proteins controlling cellular energetics. Crucially, a new role for CFTR as a kinase controller is proposed with ionic concentration acting as a signal. The model posits a regulatory control relay for energy sensing involving a cascade of protein kinases bound to CFTR.
Collapse
Affiliation(s)
- Anil Mehta
- Department of Maternal and Child Health Sciences, University of Dundee, Ninewells Hospital, Dundee DD1 9SY, UK.
| |
Collapse
|
38
|
|