1
|
Anjum R, Clarke VRJ, Nagasawa Y, Murakoshi H, Paradis S. Rem2 interacts with CaMKII at synapses and restricts long-term potentiation in hippocampus. PLoS One 2024; 19:e0301063. [PMID: 38995900 PMCID: PMC11244776 DOI: 10.1371/journal.pone.0301063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 06/11/2024] [Indexed: 07/14/2024] Open
Abstract
Synaptic plasticity, the process whereby neuronal connections are either strengthened or weakened in response to stereotyped forms of stimulation, is widely believed to represent the molecular mechanism that underlies learning and memory. The holoenzyme calcium/calmodulin-dependent protein kinase II (CaMKII) plays a well-established and critical role in the induction of a variety of forms of synaptic plasticity such as long-term potentiation (LTP), long-term depression (LTD) and depotentiation. Previously, we identified the GTPase Rem2 as a potent, endogenous inhibitor of CaMKII. Here, we report that knock out of Rem2 enhances LTP at the Schaffer collateral to CA1 synapse in hippocampus, consistent with an inhibitory action of Rem2 on CaMKII in vivo. Further, re-expression of WT Rem2 rescues the enhanced LTP observed in slices obtained from Rem2 conditional knock out (cKO) mice, while expression of a mutant Rem2 construct that is unable to inhibit CaMKII in vitro fails to rescue increased LTP. In addition, we demonstrate that CaMKII and Rem2 interact in dendritic spines using a 2pFLIM-FRET approach. Taken together, our data lead us to propose that Rem2 serves as a brake on synaptic potentiation via inhibition of CaMKII activity. Further, the enhanced LTP phenotype we observe in Rem2 cKO slices reveals a previously unknown role for Rem2 in the negative regulation of CaMKII function.
Collapse
Affiliation(s)
- Rabia Anjum
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, Massachusetts, United States of America
| | - Vernon R. J. Clarke
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Yutaro Nagasawa
- Department of Physiological Sciences, The Graduate University for Advanced Studies; Hayama, Kanagawa, Japan
- Supportive Center for Brain Research, National Institute for Physiological Sciences; Okazaki, Aichi, Japan
| | - Hideji Murakoshi
- Department of Physiological Sciences, The Graduate University for Advanced Studies; Hayama, Kanagawa, Japan
- Supportive Center for Brain Research, National Institute for Physiological Sciences; Okazaki, Aichi, Japan
| | - Suzanne Paradis
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, Massachusetts, United States of America
| |
Collapse
|
2
|
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: 2] [Impact Index Per Article: 2.0] [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
|
3
|
Zhou J, Xu B, He F, Shu Y, Chen X, Liu Z, Sun B, Zhang W. Association of RASGRP1 polymorphism with vascular complications in Chinese diabetic patients with glycemic control and antihypertensive treatment. Cardiovasc Diabetol 2024; 23:166. [PMID: 38730425 PMCID: PMC11088008 DOI: 10.1186/s12933-024-02267-2] [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: 01/22/2024] [Accepted: 05/06/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Studies have shown that RASGRP1 was potently associated with the onset of type 2 diabetes mellitus (T2DM), and RASGRP1 rs7403531 was significantly correlated with islet function in T2DM patients. However, the effect of RASGRP1 polymorphism on blood glucose and blood pressure in T2DM patients after continuous treatment has yet to be fully elucidated. OBJECTIVE This study aimed to explore the association between RASGRP1 genetic polymorphism and cardiovascular complications in T2DM patients, so as to provide more evidence for the individualized treatment of T2DM patients. METHODS We retrospectively analyzed a large-scale multicenter drug clinical study cohort that based on a 2 × 2 factorial (glucose control axis and blood pressure lowering axis) randomized controlled design, with follow-up for 5 years. The major vascular endpoint events included cardiovascular death, non-fatal stroke, coronary heart disease, new-onset or worsening renal disease, and diabetic retinopathy. RASGRP1 rs12593201, rs56254815 and rs7403531 were finally selected as candidate single nucleotide polymorphisms. Mixed linear model and Cox hazard ratio (HR) model were used for data analysis with IBM SPSS (version 20.0 for windows; Chicago, IL). RESULTS Our study enrolled 1357 patients with high-risk diabetes, with a mean follow-up duration of 4.8 years. RASGRP1 rs7403531 was associated with vascular events in hypoglycemic and antihypertensive therapy. Specifically, compared with CC carriers, patients with CT/TT genotype had fewer major microvascular events (HR = 0.41, 95% confidence interval (CI) 0.21-0.80, P = 0.009), and reduced the risk of major eye disease events (HR = 0.44, 95% CI 0.20-0.94, P = 0.03). For glucose lowering axis, CT/TT carriers had a lower risk of secondary nephropathy (HR = 0.48, 95% CI 0.25-0.92, P = 0.03) in patients with standard glycemic control. For blood pressure lowering axis, all cerebrovascular events (HR = 2.24, 95% CI 1.11-4.51, P = 0.025) and stroke events (HR = 2.07, 95% CI 1.03-4.15, P = 0.04) were increased in patients with CC genotype compared to those with CT/TT genotype in the placebo group, respectively. Furthermore, patients with CC genotype showed a reduced risk of major cerebrovascular events in antihypertensive group (HR = 0.36, 95% CI 0.15-0.86, P = 0.021). For RASGRP1 rs56254815, compared with the AA genotype carriers, the systolic blood pressure of AG/GG carriers in the antihypertensive group decreased by 1.5mmhg on average (P = 0.04). In the placebo group, the blood pressure of AG/GG carriers was 1.7mmHg higher than that of AA carriers (P = 0.02). CONCLUSION We found that patients with G allele of RASGRP1 (rs56254815) showed a better antihypertensive therapy efficacy in T2DM patients. The rs7403531 T allele could reduce the risk of major microvascular events and major eye diseases in T2DM patients receiving either hypoglycemic or antihypertensive therapy. Our findings suggest that RASGRP1 genetic polymorphism might predict the cardiovascular complications in T2DM patients.
Collapse
Affiliation(s)
- Jiecan Zhou
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, China.
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, China.
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 110 Xiangya Rode, Kaifu district, 410008, Changsha, Hunan, P.R. China.
| | - Bo Xu
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, China
| | - Fazhong He
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 110 Xiangya Rode, Kaifu district, 410008, Changsha, Hunan, P.R. China
- Pharmacogenetics Research Institute, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, 410078, Changsha, Hunan, China
- Department of Pharmacy-Quality control section of medical department, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, Guangdong, China
| | - Yan Shu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, USA
| | - Xiaoping Chen
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, China
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 110 Xiangya Rode, Kaifu district, 410008, Changsha, Hunan, P.R. China
- Pharmacogenetics Research Institute, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, 410078, Changsha, Hunan, China
| | - Zhaoqian Liu
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, China
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 110 Xiangya Rode, Kaifu district, 410008, Changsha, Hunan, P.R. China
- Pharmacogenetics Research Institute, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, 410078, Changsha, Hunan, China
| | - Bao Sun
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 110 Xiangya Rode, Kaifu district, 410008, Changsha, Hunan, P.R. China.
- Pharmacogenetics Research Institute, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, 410078, Changsha, Hunan, China.
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, People's Middle Street, Changsha, 410011, Hunan , P. R. China.
| | - Wei Zhang
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, China.
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 110 Xiangya Rode, Kaifu district, 410008, Changsha, Hunan, P.R. China.
- Pharmacogenetics Research Institute, Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, 410078, Changsha, Hunan, China.
| |
Collapse
|
4
|
Anjum R, Clarke VRJ, Nagasawa Y, Murakoshi H, Paradis S. Rem2 interacts with CaMKII at synapses and restricts long-term potentiation in hippocampus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.11.584540. [PMID: 38558974 PMCID: PMC10979978 DOI: 10.1101/2024.03.11.584540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Synaptic plasticity, the process whereby neuronal connections are either strengthened or weakened in response to stereotyped forms of stimulation, is widely believed to represent the molecular mechanism that underlies learning and memory. The holoenzyme CaMKII plays a well-established and critical role in the induction of a variety of forms of synaptic plasticity such as long-term potentiation (LTP), long-term depression (LTD) and depotentiation. Previously, we identified the GTPase Rem2 as a potent, endogenous inhibitor of CaMKII. Here, we report that knock out of Rem2 enhances LTP at the Schaffer collateral to CA1 synapse in hippocampus, consistent with an inhibitory action of Rem2 on CaMKII in vivo. Further, re-expression of WT Rem2 rescues the enhanced LTP observed in slices obtained from Rem2 conditional knock out (cKO) mice, while expression of a mutant Rem2 construct that is unable to inhibit CaMKII in vitro fails to rescue increased LTP. In addition, we demonstrate that CaMKII and Rem2 interact in dendritic spines using a 2pFLIM-FRET approach. Taken together, our data lead us to propose that Rem2 serves as a brake on runaway synaptic potentiation via inhibition of CaMKII activity. Further, the enhanced LTP phenotype we observe in Rem2 cKO slices reveals a previously unknown role for Rem2 in the negative regulation of CaMKII function.
Collapse
Affiliation(s)
- Rabia Anjum
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, Massachusetts 02454, United States of America
| | - Vernon R J Clarke
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Yutaro Nagasawa
- Department of Physiological Sciences, The Graduate University for Advanced Studies; Hayama, Kanagawa 240-0193, Japan
- Supportive Center for Brain Research, National Institute for Physiological Sciences; Okazaki, Aichi 444-8585, Japan
| | - Hideji Murakoshi
- Department of Physiological Sciences, The Graduate University for Advanced Studies; Hayama, Kanagawa 240-0193, Japan
- Supportive Center for Brain Research, National Institute for Physiological Sciences; Okazaki, Aichi 444-8585, Japan
| | - Suzanne Paradis
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, Massachusetts 02454, United States of America
| |
Collapse
|
5
|
Sun Z, Li Y, Tan X, Liu W, He X, Pan D, Li E, Xu L, Long L. Friend or Foe: Regulation, Downstream Effectors of RRAD in Cancer. Biomolecules 2023; 13:biom13030477. [PMID: 36979412 PMCID: PMC10046484 DOI: 10.3390/biom13030477] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
Ras-related associated with diabetes (RRAD), a member of the Ras-related GTPase superfamily, is primarily a cytosolic protein that actives in the plasma membrane. RRAD is highly expressed in type 2 diabetes patients and as a biomarker of congestive heart failure. Mounting evidence showed that RRAD is important for the progression and metastasis of tumor cells, which play opposite roles as an oncogene or tumor suppressor gene depending on cancer and cell type. These findings are of great significance, especially given that relevant molecular mechanisms are being discovered. Being regulated in various pathways, RRAD plays wide spectrum cellular activity including tumor cell division, motility, apoptosis, and energy metabolism by modulating tumor-related gene expression and interacting with multiple downstream effectors. Additionally, RRAD in senescence may contribute to its role in cancer. Despite the twofold characters of RRAD, targeted therapies are becoming a potential therapeutic strategy to combat cancers. This review will discuss the dual identity of RRAD in specific cancer type, provides an overview of the regulation and downstream effectors of RRAD to offer valuable insights for readers, explore the intracellular role of RRAD in cancer, and give a reference for future mechanistic studies.
Collapse
Affiliation(s)
- Zhangyue Sun
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Institute of Basic Medical Science, Shantou University Medical College, Shantou 515041, China
| | - Yongkang Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Institute of Basic Medical Science, Shantou University Medical College, Shantou 515041, China
| | - Xiaolu Tan
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Institute of Basic Medical Science, Shantou University Medical College, Shantou 515041, China
| | - Wanyi Liu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Institute of Basic Medical Science, Shantou University Medical College, Shantou 515041, China
| | - Xinglin He
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Institute of Basic Medical Science, Shantou University Medical College, Shantou 515041, China
| | - Deyuan Pan
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Institute of Basic Medical Science, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, China
| | - Enmin Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Institute of Basic Medical Science, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, China
| | - Liyan Xu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Institute of Basic Medical Science, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, China
| | - Lin Long
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Institute of Basic Medical Science, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, China
- Correspondence: ; Tel.: +86-754-88900460; Fax: +86-754-88900847
| |
Collapse
|
6
|
Phosphohistidine signaling promotes FAK-RB1 interaction and growth factor-independent proliferation of esophageal squamous cell carcinoma. Oncogene 2023; 42:449-460. [PMID: 36513743 DOI: 10.1038/s41388-022-02568-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 11/08/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022]
Abstract
Current clinical therapies targeting receptor tyrosine kinases including focal adhesion kinase (FAK) have had limited or no effect on esophageal squamous cell carcinoma (ESCC). Unlike esophageal adenocarcinomas, ESCC acquire glucose in excess of their anabolic need. We recently reported that glucose-induced growth factor-independent proliferation requires the phosphorylation of FAKHis58. Here, we confirm His58 phosphorylation in FAK immunoprecipitates of glucose-stimulated, serum-starved ESCC cells using antibodies specific for 3-phosphohistidine and mass spectrometry. We also confirm a role for the histidine kinase, NME1, in glucose-induced FAKpoHis58 and ESCC cell proliferation, correlating with increased levels of NME1 in ESCC tumors versus normal esophageal tissues. Unbiased screening identified glucose-induced retinoblastoma transcriptional corepressor 1 (RB1) binding to FAK, mediated through a "LxCxE" RB1-binding motif in FAK's FERM domain. Importantly, in the absence of growth factors, glucose increased FAK scaffolding of RB1 in the cytoplasm, correlating with increased ESCC G1→S phase transition. Our data strongly suggest that this glucose-mediated mitogenic pathway is novel and represents a unique targetable opportunity in ESCC.
Collapse
|
7
|
Functional diversity in the RAS subfamily of small GTPases. Biochem Soc Trans 2022; 50:921-933. [PMID: 35356965 DOI: 10.1042/bst20211166] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/15/2022] [Accepted: 03/21/2022] [Indexed: 12/12/2022]
Abstract
RAS small GTPases regulate important signalling pathways and are notorious drivers of cancer development and progression. While most research to date has focused on understanding and addressing the oncogenic potential of three RAS oncogenes: HRAS, KRAS, and NRAS; the full RAS subfamily is composed of 35 related GTPases with diverse cellular functions. Most remain deeply understudied despite strong evolutionary conservation. Here, we highlight a group of 17 poorly characterized RAS GTPases that are frequently down-regulated in cancer and evidence suggests may function not as oncogenes, but as tumour suppressors. These GTPases remain largely enigmatic in terms of their cellular function, regulation, and interaction with effector proteins. They cluster within two families we designate as 'distal-RAS' (D-RAS; comprised of DIRAS, RASD, and RASL10) and 'CaaX-Less RAS' (CL-RAS; comprised of RGK, NKIRAS, RERG, and RASL11/12 GTPases). Evidence of a tumour suppressive role for many of these GTPases supports the premise that RAS subfamily proteins may collectively regulate cellular proliferation.
Collapse
|
8
|
Spontaneous Cell Detachment and Reattachment in Cancer Cell Lines: An In Vitro Model of Metastasis and Malignancy. Int J Mol Sci 2021; 22:ijms22094929. [PMID: 34066490 PMCID: PMC8124513 DOI: 10.3390/ijms22094929] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/27/2021] [Accepted: 04/30/2021] [Indexed: 11/16/2022] Open
Abstract
There is an unmet need for simplified in vitro models of malignancy and metastasis that facilitate fast, affordable and scalable gene and compound analysis. "Adherent" cancer cell lines frequently release "free-floating" cells into suspension that are viable and can reattach. This, in a simplistic way, mimics the metastatic process. We compared the gene expression profiles of naturally co-existing populations of floating and adherent cells in SW620 (colon), C33a (cervix) and HeLa (cervix) cancer cells. We found that 1227, 1367 and 1333 genes were at least 2-fold differentially expressed in the respective cell lines, of which 122 were shared among the three cell lines. As proof of principle, we focused on the anti-metastatic gene NM23-H1, which was downregulated both at the RNA and protein level in the floating cell populations of all three cell lines. Knockdown of NM23-H1 significantly increased the number of floating (and viable) cells, whereas overexpression of NM23-H1 significantly reduced the proportion of floating cells. Other potential regulators of these cellular states were identified through pathway analysis, including hypoxia, mTOR (mechanistic target of rapamycin), cell adhesion and cell polarity signal transduction pathways. Hypoxia, a condition linked to malignancy and metastasis, reduced NM23-H1 expression and significantly increased the number of free-floating cells. Inhibition of mTOR or Rho-associated protein kinase (ROCK) significantly increased cell death specifically in the floating and not the adherent cell population. In conclusion, our study suggests that dynamic subpopulations of free-floating and adherent cells is a useful model to screen and identify genes, drugs and pathways that regulate the process of cancer metastasis, such as cell detachment and anoikis.
Collapse
|
9
|
Activation of Nm23-H1 to suppress breast cancer metastasis via redox regulation. Exp Mol Med 2021; 53:346-357. [PMID: 33753879 PMCID: PMC8080780 DOI: 10.1038/s12276-021-00575-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/21/2020] [Accepted: 01/12/2021] [Indexed: 02/05/2023] Open
Abstract
Non-metastatic protein 23 H1 (Nm23-H1), a housekeeping enzyme, is a nucleoside diphosphate kinase-A (NDPK-A). It was the first identified metastasis suppressor protein. Nm23-H1 prolongs disease-free survival and is associated with a good prognosis in breast cancer patients. However, the molecular mechanisms underlying the role of Nm23-H1 in biological processes are still not well understood. This is a review of recent studies focusing on controlling NDPK activity based on the redox regulation of Nm23-H1, structural, and functional changes associated with the oxidation of cysteine residues, and the relationship between NDPK activity and cancer metastasis. Further understanding of the redox regulation of the NDPK function will likely provide a new perspective for developing new strategies for the activation of NDPK-A in suppressing cancer metastasis.
Collapse
|
10
|
Filić V, Marinović M, Šoštar M, Weber I. Modulation of small GTPase activity by NME proteins. J Transl Med 2018; 98:589-601. [PMID: 29434248 DOI: 10.1038/s41374-018-0023-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 12/06/2017] [Accepted: 12/29/2017] [Indexed: 12/26/2022] Open
Abstract
NME proteins are reported to influence signal transduction activity of small GTPases from the Ras superfamily by diverse mechanisms in addition to their generic NDP kinase activity, which replenishes the cytoplasmic pool of GTP. Comprehensive evidence shows that NME proteins modulate the activity of Ras GTPases, in particular members of the Rho family, via binding to their major activators GEFs. Direct interaction between several NMEs and Ras GTPases were also indicated in vitro and in vivo. These modes of regulation are mainly independent of the NME's kinase activity. NMEs also modulate the Ras-mediated signal transduction by interfering with the formation of a Ras signaling complex at the plasma membrane. In several examples, NMEs were proposed to perform the role of GAP proteins by promoting hydrolysis of the bound GTP, but this activity still requires additional verification. Early suggestions that NMEs can activate small GTPases by direct phosphorylation of the bound GDP, or by high-rate loading of GTP onto a closely apposed GTPase, were largely dismissed. In this review article, we survey and put into perspective published examples of identified and hypothetical mechanisms of Ras signaling modulation by NME proteins. We also point out involvement of NMEs in the transcriptional regulation of components of Ras GTPases-mediated signal transduction pathways, and reciprocal regulation of NME function by small GTPases, particularly related to NME's binding to membranes.
Collapse
Affiliation(s)
- Vedrana Filić
- Ruđer Bošković Institute, Division of Molecular Biology, Bijenička 54, HR-10000, Zagreb, Croatia
| | - Maja Marinović
- Ruđer Bošković Institute, Division of Molecular Biology, Bijenička 54, HR-10000, Zagreb, Croatia
| | - Marko Šoštar
- Ruđer Bošković Institute, Division of Molecular Biology, Bijenička 54, HR-10000, Zagreb, Croatia
| | - Igor Weber
- Ruđer Bošković Institute, Division of Molecular Biology, Bijenička 54, HR-10000, Zagreb, Croatia.
| |
Collapse
|
11
|
Abstract
Nucleoside diphosphate kinases (NDPK) are nucleotide metabolism enzymes encoded by NME genes (also called NM23). Given the fact that not all NME-encoded proteins are catalytically active NDPKs and that NM23 generally refers to clinical studies on metastasis, we use here NME/NDPK to denote the proteins. Since their discovery in the 1950's, NMEs/NDPKs have been shown to be involved in multiple physiological and pathological cellular processes, but the molecular mechanisms have not been fully determined. Recent progress in elucidating these underlying mechanisms has been presented by experts in the field at the 10th International Congress on the NDPK/NME/AWD protein family in October 2016 in Dubrovnik, Croatia, and is summarized in review articles or original research in this and an upcoming issue of Laboratory Investigation. Within this editorial, we discuss three major cellular processes that involve members of the multi-functional NME/NDPK family: (i) cancer and metastasis dissemination, (ii) membrane remodeling and nucleotide channeling, and iii) protein histidine phosphorylation.
Collapse
|
12
|
Oncogenic Epstein-Barr virus recruits Nm23-H1 to regulate chromatin modifiers. J Transl Med 2018; 98:258-268. [PMID: 29035376 PMCID: PMC6053075 DOI: 10.1038/labinvest.2017.112] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 08/03/2017] [Accepted: 08/16/2017] [Indexed: 12/17/2022] Open
Abstract
In cancer progression, metastasis is a major cause of poor survival of patients and can be targeted for therapeutic interventions. The first discovered metastatic-suppressor Nm23-H1 possesses nucleoside diphosphate kinase, histidine kinase, and DNase activity as a broad-spectrum enzyme. Recent advances in cancer metastasis have opened new ways for the development of therapeutic molecular approaches. In this review, we provide a summary of the current understanding of Nm23/NDPKs in the context of viral oncogenesis. We also focused on Nm23-H1-mediated cellular events with an emphasis on chromatin modifications. How Nm23-H1 modulates the activities of chromatin modifiers through interaction with Epstein-Barr virus-encoded oncogenic antigens and related crosstalks are discussed in the context of other oncogenic viruses. We also described the current understanding of the cellular and viral interactions of Nm23-H1 and their reference to transcription regulation and metastasis. Further, we summarized the recent therapeutic approaches targeting Nm23 and its potential links to pathways that can be exploited by oncogenic viruses.
Collapse
|
13
|
Kenny K, Royer L, Moore AR, Chen X, Marr MT, Paradis S. Rem2 signaling affects neuronal structure and function in part by regulation of gene expression. Mol Cell Neurosci 2017; 85:190-201. [PMID: 29066292 DOI: 10.1016/j.mcn.2017.10.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 09/27/2017] [Accepted: 10/11/2017] [Indexed: 12/12/2022] Open
Abstract
The central nervous system has the remarkable ability to convert changes in the environment in the form of sensory experience into long-term alterations in synaptic connections and dendritic arborization, in part through changes in gene expression. Surprisingly, the molecular mechanisms that translate neuronal activity into changes in neuronal connectivity and morphology remain elusive. Rem2, a member of the Rad/Rem/Rem2/Gem/Kir (RGK) subfamily of small Ras-like GTPases, is a positive regulator of synapse formation and negative regulator of dendritic arborization. Here we identify that one output of Rem2 signaling is the regulation of gene expression. Specifically, we demonstrate that Rem2 signaling modulates the expression of genes required for a variety of cellular processes from neurite extension to synapse formation and synaptic function. Our results highlight Rem2 as a unique molecule that transduces changes in neuronal activity detected at the cell membrane to morphologically relevant changes in gene expression in the nucleus.
Collapse
Affiliation(s)
- Katelyn Kenny
- Department of Biology, Brandeis University, Waltham, MA 02454, United States
| | - Leandro Royer
- Department of Biology, Brandeis University, Waltham, MA 02454, United States
| | - Anna R Moore
- Department of Biology, Brandeis University, Waltham, MA 02454, United States; Volen Center for Complex Systems, Brandeis University, Waltham, MA 02454, United States; National Center for Behavioral Genomics, Brandeis University, Waltham, MA 02454, United States
| | - Xiao Chen
- Department of Biology, Brandeis University, Waltham, MA 02454, United States; National Center for Behavioral Genomics, Brandeis University, Waltham, MA 02454, United States
| | - Michael T Marr
- Department of Biology, Brandeis University, Waltham, MA 02454, United States; Rosenstiel Basic Medical Sciences Research Center, Brandeis University, Waltham, MA 02454, United States
| | - Suzanne Paradis
- Department of Biology, Brandeis University, Waltham, MA 02454, United States; Volen Center for Complex Systems, Brandeis University, Waltham, MA 02454, United States; National Center for Behavioral Genomics, Brandeis University, Waltham, MA 02454, United States.
| |
Collapse
|
14
|
Chang DD, Colecraft HM. Rad and Rem are non-canonical G-proteins with respect to the regulatory role of guanine nucleotide binding in Ca(V)1.2 channel regulation. J Physiol 2016; 593:5075-90. [PMID: 26426338 DOI: 10.1113/jp270889] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 09/27/2015] [Indexed: 12/15/2022] Open
Abstract
Rad and Rem are Ras-like G-proteins linked to diverse cardiovascular functions and pathophysiology. Understanding how Rad and Rem are regulated is important for deepened insights into their pathophysiological roles. As in other Ras-like G-proteins, Rad and Rem contain a conserved guanine-nucleotide binding domain (G-domain). Canonically, G-domains are key control modules, functioning as nucleotide-regulated switches of G-protein activity. Whether Rad and Rem G-domains conform to this canonical paradigm is ambiguous. Here, we used multiple functional measurements in HEK293 cells and cardiomyocytes (Ca(V)1.2 currents, Ca(2+) transients, Ca(V)β binding) as biosensors to probe the role of the G-domain in regulation of Rad and Rem function. We utilized Rad(S105N) and Rem(T94N), which are the cognate mutants to Ras(S17N), a dominant-negative variant of Ras that displays decreased nucleotide binding affinity. In HEK293 cells, over-expression of either Rad(S105N) or Rem(T94N) strongly inhibited reconstituted Ca(V)1.2 currents to the same extent as their wild-type (wt) counterparts, contrasting with reports that Rad(S105N) is functionally inert in HEK293 cells. Adenovirus-mediated expression of either wt Rad or Rad(S105N) in cardiomyocytes dramatically blocked L-type calcium current (I(Ca,L)) and inhibited Ca(2+)-induced Ca(2+) release, contradicting reports that Rad(S105N) acts as a dominant negative in heart. By contrast, Rem(T94N) was significantly less effective than wt Rem at inhibiting I(Ca,L) and Ca(2+) transients in cardiomyocytes. FRET analyses in cardiomyocytes revealed that both Rad(S105N) and Rem(T94N) had moderately reduced binding affinity for Ca(V)βs relative to their wt counterparts. The results indicate Rad and Rem are non-canonical G-proteins with respect to the regulatory role of their G-domain in Ca(V)1.2 regulation.
Collapse
Affiliation(s)
- Donald D Chang
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, 10032, USA
| | - Henry M Colecraft
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, 10032, USA
| |
Collapse
|
15
|
You DJ, Park CR, Mander S, Ahn C, Seong JY, Hwang JI. Characterization of Functional Domains in NME1L Regulation of NF-κB Signaling. Mol Cells 2016; 39:403-9. [PMID: 27094059 PMCID: PMC4870188 DOI: 10.14348/molcells.2016.2320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 02/24/2016] [Accepted: 02/29/2016] [Indexed: 11/27/2022] Open
Abstract
NME1 is a well-known metastasis suppressor which has been reported to be downregulated in some highly aggressive cancer cells. Although most studies have focused on NME1, the NME1 gene also encodes the protein (NME1L) containing N-terminal 25 extra amino acids by alternative splicing. According to previous studies, NME1L has potent anti-metastatic activity, in comparison with NME1, by interacting with IKKβ and regulating its activity. In the present study, we tried to define the role of the N-terminal 25 amino acids of NME1L in NF-κB activation signaling. Unfortunately, the sequence itself did not interact with IKKβ, suggesting that it may be not enough to constitute the functional structure. Further construction of NME1L fragments and biochemical analysis revealed that N-terminal 84 residues constitute minimal structure for homodimerization, IKKβ interaction and regulation of NF-κB signaling. The inhibitory effect of the fragment on cancer cell migration and NF-κB-stimulated gene expression was equivalent to that of whole NME1L. The data suggest that the N-terminal 84 residues may be a core region for the anti-metastatic activity of NME1L. Based on this result, further structural analysis of the binding between NME1L and IKKβ may help in understanding the anti-metastatic activity of NME1L and provide direction to NME1L and IKKβ-related anti-cancer drug design.
Collapse
Affiliation(s)
- Dong-Joo You
- Graduate School of Medicine, Korea University, Seoul 136-705,
Korea
| | - Cho Rong Park
- Graduate School of Medicine, Korea University, Seoul 136-705,
Korea
| | - Sunam Mander
- Graduate School of Medicine, Korea University, Seoul 136-705,
Korea
| | - Curie Ahn
- Transplantation Research Institute, Cancer Research Institute, Seoul National University, Seoul 110-799,
Korea
| | - Jae Young Seong
- Graduate School of Medicine, Korea University, Seoul 136-705,
Korea
| | - Jong-Ik Hwang
- Graduate School of Medicine, Korea University, Seoul 136-705,
Korea
| |
Collapse
|
16
|
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
|
17
|
You D, Mander S, Park CR, Koo O, Lee C, Oh S, Ahn C, Seong JY, Hwang J. NME1L Negatively Regulates IGF1‐Dependent Proliferation of Breast Cancer Cells. J Cell Biochem 2015; 117:1454-63. [DOI: 10.1002/jcb.25441] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 11/11/2015] [Indexed: 12/15/2022]
Affiliation(s)
- Dong‐Joo You
- Graduate School of MedicineKorea University, 73 Inchon‐ro, Seongbuk‐guSeoul136‐705Republic of Korea
| | - Sunam Mander
- Graduate School of MedicineKorea University, 73 Inchon‐ro, Seongbuk‐guSeoul136‐705Republic of Korea
| | - Cho Rong Park
- Graduate School of MedicineKorea University, 73 Inchon‐ro, Seongbuk‐guSeoul136‐705Republic of Korea
| | - Okjae Koo
- Samsung Biomedical Research Institute130 Samsung‐ro, Yeongtong‐gu, Suwon‐siGyeonggi‐do433‐803Republic of Korea
| | - Cheolju Lee
- Life Sciences DivisionKorea Institute of Science and Technology, Seongbuk‐guSeoul136‐791Republic of Korea
| | - Seong‐Hyun Oh
- College of PharmacyGachon UniversityIncheon406‐840Republic of Korea
| | - Curie Ahn
- Transplantation Research InstituteCancer Research Institute, Seoul National University, Yongun‐dong, Jongno‐guSeoul110‐799Republic of Korea
| | - Jae Young Seong
- Graduate School of MedicineKorea University, 73 Inchon‐ro, Seongbuk‐guSeoul136‐705Republic of Korea
| | - Jong‐Ik Hwang
- Graduate School of MedicineKorea University, 73 Inchon‐ro, Seongbuk‐guSeoul136‐705Republic of Korea
| |
Collapse
|
18
|
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: 1.7] [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
|
19
|
The ubiquitin E3 ligase SCF-FBXO24 recognizes deacetylated nucleoside diphosphate kinase A to enhance its degradation. Mol Cell Biol 2015; 35:1001-13. [PMID: 25582197 DOI: 10.1128/mcb.01185-14] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Skp-Cul-F box (SCF) ubiquitin E3 ligase machinery recognizes predominantly phosphodegrons or, less commonly, an (I/L)Q molecular signature within substrates to facilitate their recruitment in mediating protein ubiquitination and degradation. Here, we examined the molecular signals that determine the turnover of the multifunctional enzyme nucleoside diphosphate kinase A (NDPK-A) that controls cell proliferation. NDPK-A protein exhibits a half-life of ∼6 h in HeLa cells and is targeted for ubiquitylation through actions of the F-box protein FBXO24. SCF-FBXO24 polyubiquitinates NDPK-A at K85, and two NH(2)-terminal residues, L55 and K56, were identified as important molecular sites for FBXO24 interaction. Importantly, K56 acetylation impairs its interaction with FBXO24, and replacing K56 with Q56, an acetylation mimic, reduces NDPK-A FBXO24 binding capacity. The acetyltransferase GCN5 catalyzes K56 acetylation within NDPK-A, thereby stabilizing NDPK-A, whereas GCN5 depletion in cells accelerates NDPK-A degradation. Cellular expression of an NDPK-A acetylation mimic or FBXO24 silencing increases NDPK-A life span which, in turn, impairs cell migration and wound healing. We propose that lysine acetylation when presented in the appropriate context may be recognized by some F-box proteins as a unique inhibitory molecular signal for their recruitment to restrict substrate degradation.
Collapse
|
20
|
Janus-faces of NME-oncoprotein interactions. Naunyn Schmiedebergs Arch Pharmacol 2014; 388:175-87. [PMID: 25366701 DOI: 10.1007/s00210-014-1062-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 10/15/2014] [Indexed: 12/26/2022]
Abstract
Since the identification of Nm23 (NME1, NME/NM23 nucleoside diphosphate kinase 1) as the first non-metastatic protein, a great deal of research on members of the NME family of proteins has focused on roles in processes implicated in carcinogenesis and particularly their regulation of cellular motility and the process of metastatic spread. To date, there are ten identified members of this family of genes, and these can be dichotomized into groups both taxonomically and by the presence or absence of their nucleoside diphosphate kinase activity with NMEs 1-4 encoding nucleoside diphosphate kinases (NDPKs) and NMEs 5-9 plus RP2 displaying little if any NDPK activity. NMEs are relatively small proteins that can form hetero-oligomers (typically hexamers), and given the apparent genetic redundancy of some NMEs and the number of different isoforms, it is perhaps not surprising that there remains a great deal of uncertainty regarding their function and even more regarding cellular mechanisms of action. Since residues that contribute to NDPK activity span much of the protein, it seems likely that the consequences of NME expression must be mediated through their NDPK activity, through interactions with other structures in cells including protein-protein interactions or through combinations of these. Our goal in this review is to focus on some of the protein-protein interactions that have been identified and to highlight some of the challenges that face this area of research.
Collapse
|
21
|
Banerjee S, Jha HC, Robertson ES. Regulation of the metastasis suppressor Nm23-H1 by tumor viruses. Naunyn Schmiedebergs Arch Pharmacol 2014; 388:207-24. [PMID: 25199839 DOI: 10.1007/s00210-014-1043-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 08/21/2014] [Indexed: 12/16/2022]
Abstract
Metastasis is the most common cause of cancer mortality. To increase the survival of patients, it is necessary to develop more effective methods for treating as well as preventing metastatic diseases. Recent advancement of knowledge in cancer metastasis provides the basis for development of targeted molecular therapeutics aimed at the tumor cell or its interaction with the host microenvironment. Metastasis suppressor genes (MSGs) are promising targets for inhibition of the metastasis process. During the past decade, functional significance of these genes, their regulatory pathways, and related downstream effector molecules have become a major focus of cancer research. Nm23-H1, first in the family of Nm23 human homologues, is a well-characterized, anti-metastatic factor linked with a large number of human malignancies. Mounting evidence to date suggests an important role for Nm23-H1 in reducing virus-induced tumor cell motility and migration. A detailed understanding of the molecular association between oncogenic viral antigens with Nm23-H1 may reveal the underlying mechanisms for tumor virus-associated malignancies. In this review, we will focus on the recent advances to our understanding of the molecular basis of oncogenic virus-induced progression of tumor metastasis by deregulation of Nm23-H1.
Collapse
Affiliation(s)
- Shuvomoy Banerjee
- Department of Microbiology and Tumor Virology Program, Abramson Comprehensive Cancer Center, Perelman School of Medicine at the University of Pennsylvania, 201E Johnson Pavilion, 3610 Hamilton Walk, Philadelphia, PA, 19104, USA
| | | | | |
Collapse
|
22
|
Li HR, Huang LS, Chen G, Lu HP, Feng ZB. Correlation between GPC3 expression and proliferation of hepatocellular carcinoma cells. Shijie Huaren Xiaohua Zazhi 2014; 22:2108-2115. [DOI: 10.11569/wcjd.v22.i15.2108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the relationship between GPC3 expression and proliferative ability of hepatocellular carcinoma (HCC) cells.
METHODS: Immunohistochemistry was employed to detect the expression of GPC3 in 54 HCC tissues. The correlation between GPC3 expression and Ki-67 labeling index and other clinicopathologic characteristics was studied. An shRNA targeting the GPC3 gene was transfected into SMMC7721 cells to silence GPC3 expression. MTT assay was performed to determine the effect of GPC3 knockdown on the growth of HCC SMMC7721 cells.
RESULTS: GPC3 expression was significantly correlated with Ki-67 Li (r = 0.521, P = 0.000). GPC3 expression was related to differentiation degree and vessel invasion of HCC (r = 0.287, 0.289; P = 0.035, 0.034). The cell proliferative activity was significantly reduced in the GPC3-shRNA group compared with the negative control group and blank control group (F = 45.38, P = 0.000).
CONCLUSION: GPC3 may play an important role in the occurrence, development and invasion of HCC, mainly due to the effect on the proliferation of HCC cells.
Collapse
|
23
|
You DJ, Park CR, Lee HB, Moon MJ, Kang JH, Lee C, Oh SH, Ahn C, Seong JY, Hwang JI. A splicing variant of NME1 negatively regulates NF-κB signaling and inhibits cancer metastasis by interacting with IKKβ. J Biol Chem 2014; 289:17709-20. [PMID: 24811176 DOI: 10.1074/jbc.m114.553552] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
IKKβ functions as a principal upstream activator of the canonical NF-κB pathway by phosphorylating IκB, leading to its proteasomal degradation. Because IKKβ is considered a therapeutic target, understanding its regulation may facilitate the design of efficient regulators of this molecule. Here, we report a novel IKKβ-interacting molecule, NME1L, a splicing variant of the NME1 protein. NME1 has attracted attention in cancer research because of its antimetastatic activity and reduced expression in multiple aggressive types of cancer. However, the effect was just moderate but not dramatic in anti-cancer activities. We found that only NME1L interacts with IKKβ. Exogenous expression of NME1L resulted in a potent decrease in TNFα-stimulated NF-κB activation, whereas knockdown of NME1/NME1L with shRNA enhanced activity of NF-κB. NME1L down-regulates IKKβ signaling by blocking IKKβ-mediated IκB degradation. When NME1L was introduced into highly metastatic HT1080 cells, the mobility was efficiently inhibited. Furthermore, in a metastasis assay, NME1L-expressing cells did not colonize the lung. Based on these results, NME1L is a potent antimetastatic protein and may be a useful weapon in the fight against cancers.
Collapse
Affiliation(s)
- Dong-Joo You
- From the Graduate School of Medicine, Korea University, 73 Inchon-ro, Seongbuk-gu, Seoul 136-705, Korea
| | - Cho Rong Park
- From the Graduate School of Medicine, Korea University, 73 Inchon-ro, Seongbuk-gu, Seoul 136-705, Korea
| | - Hyun Bok Lee
- From the Graduate School of Medicine, Korea University, 73 Inchon-ro, Seongbuk-gu, Seoul 136-705, Korea
| | - Mi Jin Moon
- From the Graduate School of Medicine, Korea University, 73 Inchon-ro, Seongbuk-gu, Seoul 136-705, Korea
| | - Ju-Hee Kang
- the National Cancer Center, Goyang-si, Gyeonggi-do 410-769, Korea
| | - Cheolju Lee
- the Life Sciences Division, Korea Institute of Science and Technology, Seongbuk-gu, Seoul 136-791, Korea
| | - Seong-Hyun Oh
- the College of Pharmacy, Gachon University, Incheon 406-840, Korea, and
| | - Curie Ahn
- the Transplantation Research Institute, Cancer Research Institute, Seoul National University, Yongun-dong, Jongno-gu, Seoul 110-799, Korea
| | - Jae Young Seong
- From the Graduate School of Medicine, Korea University, 73 Inchon-ro, Seongbuk-gu, Seoul 136-705, Korea
| | - Jong-Ik Hwang
- From the Graduate School of Medicine, Korea University, 73 Inchon-ro, Seongbuk-gu, Seoul 136-705, Korea,
| |
Collapse
|
24
|
Niitsu N. The Association of nm23-H1 Expression with a Poor Prognosis in Patients with Peripheral T-Cell Lymphoma, Not Otherwise Specified. J Clin Exp Hematop 2014; 54:171-7. [DOI: 10.3960/jslrt.54.171] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
25
|
Marino N, Nakayama J, Collins JW, Steeg PS. Insights into the biology and prevention of tumor metastasis provided by the Nm23 metastasis suppressor gene. Cancer Metastasis Rev 2013; 31:593-603. [PMID: 22706779 DOI: 10.1007/s10555-012-9374-8] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Metastatic disease is the major cause of death among cancer patients. A class of genes, named metastasis suppressors, has been described to specifically regulate the metastatic process. The metastasis suppressor genes are downregulated in the metastatic lesion compared to the primary tumor. In this review, we describe the body of research surrounding the first metastasis suppressor identified, Nm23. Nm23 overexpression in aggressive cancer cell lines reduced their metastatic potential in vivo with no significant reduction in primary tumor size. A complex mechanism of anti-metastatic action is unfolding involving several known Nm23 enzymatic activities (nucleotide diphosphate kinase, histidine kinase, and 3'-5' exonuclease), protein-protein interactions, and downstream gene regulation properties. Translational approaches involving Nm23 have progressed to the clinic. The upregulation of Nm23 expression by medroxyprogesterone acetate has been tested in a phase II trial. Other approaches with significant preclinical success include gene therapy using traditional or nanoparticle delivery, and cell permeable Nm23 protein. Recently, based on the inverse correlation of Nm23 and LPA1 expression, a LPA1 inhibitor has been shown to both inhibit metastasis and induce metastatic dormancy.
Collapse
Affiliation(s)
- Natascia Marino
- Women's Cancers Section, Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, 37 Convent Drive, Room 1122, Bethesda, MD 20892, USA.
| | | | | | | |
Collapse
|
26
|
Yang T, Colecraft HM. Regulation of voltage-dependent calcium channels by RGK proteins. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2012; 1828:1644-54. [PMID: 23063948 DOI: 10.1016/j.bbamem.2012.10.005] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 10/04/2012] [Accepted: 10/05/2012] [Indexed: 12/28/2022]
Abstract
RGK proteins belong to the Ras superfamily of monomeric G-proteins, and currently include four members - Rad, Rem, Rem2, and Gem/Kir. RGK proteins are broadly expressed, and are the most potent known intracellular inhibitors of high-voltage-activated Ca²⁺ (Ca(V)1 and Ca(V)2) channels. Here, we review and discuss the evidence in the literature regarding the functional mechanisms, structural determinants, physiological role, and potential practical applications of RGK-mediated inhibition of Ca(V)1/Ca(V)2 channels. This article is part of a Special Issue entitled: Calcium channels.
Collapse
Affiliation(s)
- Tingting Yang
- Department of Physiology and Cellular Biophysics, Columbia University, College of Physicians and Surgeons, 1150 St. Nicholas Avenue, New York, NY 10032, USA
| | | |
Collapse
|
27
|
Marino N, Marshall JC, Steeg PS. Protein-protein interactions: a mechanism regulating the anti-metastatic properties of Nm23-H1. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2011. [PMID: 21713383 DOI: 10.07/s00210-011-0646-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Nm23-H1, also known as NDPK-A, was the first of a class of metastasis suppressor genes to be identified. Overexpression of Nm23-H1 in metastatic cell lines (melanoma, breast carcinoma, prostate, colon, hepatocellular, and oral squamous cell carcinoma) reduced cell motility in in vitro assays and metastatic potential in xenograft models, without a significant effect on primary tumor size. The mechanism of Nm23-H1 suppression of metastasis, however, is incompletely understood. Nm23-H1 has been reported to bind proteins, including those in small G-protein complexes, transcriptional complexes, the Map kinase, the TGF-β signaling pathways and the cytoskeleton. Evidence supporting these associations is presented together with evidence of resultant biochemical and phenotypic consequences of association. Cumulatively, the data suggest that part of the anti-metastatic function of Nm23-H1 lies in pathways that it interrupts via binding and inactivation of proteins.
Collapse
Affiliation(s)
- Natascia Marino
- Women's Cancers Section, Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.
| | | | | |
Collapse
|
28
|
Extracellular NM23 Protein as a Therapeutic Target for Hematologic Malignancies. Adv Hematol 2011; 2012:879368. [PMID: 21941554 PMCID: PMC3175692 DOI: 10.1155/2012/879368] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Accepted: 06/29/2011] [Indexed: 01/19/2023] Open
Abstract
An elevated serum level of NM23-H1 protein is a poor prognostic factor in patients with various hematologic malignancies. The extracellular NM23-H1 protein promotes the in vitro growth and survival of acute myelogenous leukemia (AML) cells and inversely inhibits the in vitro survival of normal peripheral blood monocytes in primary culture at concentrations equivalent to the levels found in the serum of AML patients. The growth and survival promoting activity to AML cells is associated with cytokine production and activation of mitogen-activated protein kinases (MAPKs) and signal transducers and activators of transcription (STAT) signaling pathways. Inhibitors specific for MAPK signaling pathways inhibit the growth/survival-promoting activity of NM23-H1. These findings indicate a novel biological action of extracellular NM23-H1 and its association with poor prognosis. These results suggest an important role of extracellular NM23-H1 in the malignant progression of leukemia and a potential therapeutic target for these malignancies.
Collapse
|
29
|
Saha A, Robertson ES. Functional modulation of the metastatic suppressor Nm23-H1 by oncogenic viruses. FEBS Lett 2011; 585:3174-84. [PMID: 21846466 DOI: 10.1016/j.febslet.2011.08.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 08/04/2011] [Accepted: 08/05/2011] [Indexed: 12/17/2022]
Abstract
Evidence over the last two decades from a number of disciplines has solidified some fundamental concepts in metastasis, a major contributor to cancer associated deaths. However, significant advances have been made in controlling this critical cellular process by focusing on targeted therapy. A key set of factors associated with this invasive phenotype is the nm23 family of over twenty metastasis-associated genes. Among the eight known isoforms, Nm23-H1 is the most studied potential anti-metastatic factor associated with human cancers. Importantly, a growing body of work has clearly suggested a critical role for Nm23-H1 in limiting tumor cell motility and progression induced by several tumor viruses, including Epstein-Barr virus (EBV), Kaposi's sarcoma associated herpes virus (KSHV) and human papilloma virus (HPV). A more in depth understanding of the interactions between tumor viruses encoded antigens and Nm23-H1 will facilitate the elucidation of underlying mechanism(s) which contribute to virus-associated cancers. Here, we review recent studies to explore the molecular links between human oncogenic viruses and progression of metastasis, in particular the deregulation of Nm23-H1 mediated suppression.
Collapse
Affiliation(s)
- Abhik Saha
- Department of Microbiology and Tumor Virology Program, Abramson Comprehensive Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | | |
Collapse
|
30
|
Marino N, Marshall JC, Steeg PS. Protein-protein interactions: a mechanism regulating the anti-metastatic properties of Nm23-H1. Naunyn Schmiedebergs Arch Pharmacol 2011; 384:351-62. [PMID: 21713383 DOI: 10.1007/s00210-011-0646-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Accepted: 03/14/2011] [Indexed: 01/12/2023]
Abstract
Nm23-H1, also known as NDPK-A, was the first of a class of metastasis suppressor genes to be identified. Overexpression of Nm23-H1 in metastatic cell lines (melanoma, breast carcinoma, prostate, colon, hepatocellular, and oral squamous cell carcinoma) reduced cell motility in in vitro assays and metastatic potential in xenograft models, without a significant effect on primary tumor size. The mechanism of Nm23-H1 suppression of metastasis, however, is incompletely understood. Nm23-H1 has been reported to bind proteins, including those in small G-protein complexes, transcriptional complexes, the Map kinase, the TGF-β signaling pathways and the cytoskeleton. Evidence supporting these associations is presented together with evidence of resultant biochemical and phenotypic consequences of association. Cumulatively, the data suggest that part of the anti-metastatic function of Nm23-H1 lies in pathways that it interrupts via binding and inactivation of proteins.
Collapse
Affiliation(s)
- Natascia Marino
- Women's Cancers Section, Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.
| | | | | |
Collapse
|
31
|
Proteomic analysis of NME1/NDPK A null mouse liver: evidence for a post-translational regulation of annexin IV and EF-1Bα. Naunyn Schmiedebergs Arch Pharmacol 2011; 384:407-19. [DOI: 10.1007/s00210-011-0639-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2011] [Accepted: 04/07/2011] [Indexed: 01/12/2023]
|
32
|
Hurst DR, Welch DR. Metastasis suppressor genes at the interface between the environment and tumor cell growth. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2011; 286:107-80. [PMID: 21199781 DOI: 10.1016/b978-0-12-385859-7.00003-3] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The molecular mechanisms and genetic programs required for cancer metastasis are sometimes overlapping, but components are clearly distinct from those promoting growth of a primary tumor. Every sequential, rate-limiting step in the sequence of events leading to metastasis requires coordinated expression of multiple genes, necessary signaling events, and favorable environmental conditions or the ability to escape negative selection pressures. Metastasis suppressors are molecules that inhibit the process of metastasis without preventing growth of the primary tumor. The cellular processes regulated by metastasis suppressors are diverse and function at every step in the metastatic cascade. As we gain knowledge into the molecular mechanisms of metastasis suppressors and cofactors with which they interact, we learn more about the process, including appreciation that some are potential targets for therapy of metastasis, the most lethal aspect of cancer. Until now, metastasis suppressors have been described largely by their function. With greater appreciation of their biochemical mechanisms of action, the importance of context is increasingly recognized especially since tumor cells exist in myriad microenvironments. In this chapter, we assemble the evidence that selected molecules are indeed suppressors of metastasis, collate the data defining the biochemical mechanisms of action, and glean insights regarding how metastasis suppressors regulate tumor cell communication to-from microenvironments.
Collapse
Affiliation(s)
- Douglas R Hurst
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | |
Collapse
|
33
|
Nucleoside diphosphate kinase Nm23-H1 regulates chromosomal stability by activating the GTPase dynamin during cytokinesis. Proc Natl Acad Sci U S A 2010; 107:15461-6. [PMID: 20713695 DOI: 10.1073/pnas.1010633107] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Chromosomal instability and the subsequent genetic mutations are considered to be critical factors in the development of the majority of solid tumors. Here, we describe how the nucleoside diphosphate kinase Nm23-H1, a protein with a known link to cancer progression, regulates a critical step during cytokinesis. Nm23-H1 acts to provide a local source of GTP for the GTPase dynamin. Loss of Nm23-H1 in diploid cells leads to cytokinetic furrow regression, followed by cytokinesis failure and generation of tetraploid cells. Loss of dynamin phenocopies loss of Nm23-H1, and ectopic overexpression of WT dynamin complements the loss of Nm23-H1. In the absence of p53 signaling, the tetraploid cells resulting from loss of Nm23-H1 continue cycling and develop classic hallmarks of tumor cells. We thus provide evidence that the loss of Nm23-H1, an event suspected to promote metastasis, may additionally function at an earlier stage of tumor development to drive the acquisition of chromosomal instability.
Collapse
|
34
|
Abstract
MOTIVATION An observed metabolic response is the result of the coordinated activation and interaction between multiple genetic pathways. However, the complex structure of metabolism has meant that a compete understanding of which pathways are required to produce an observed metabolic response is not fully understood. In this article, we propose an approach that can identify the genetic pathways which dictate the response of metabolic network to specific experimental conditions. RESULTS Our approach is a combination of probabilistic models for pathway ranking, clustering and classification. First, we use a non-parametric pathway extraction method to identify the most highly correlated paths through the metabolic network. We then extract the defining structure within these top-ranked pathways using both Markov clustering and classification algorithms. Furthermore, we define detailed node and edge annotations, which enable us to track each pathway, not only with respect to its genetic dependencies, but also allow for an analysis of the interacting reactions, compounds and KEGG sub-networks. We show that our approach identifies biologically meaningful pathways within two microarray expression datasets using entire KEGG metabolic networks. AVAILABILITY AND IMPLEMENTATION An R package containing a full implementation of our proposed method is currently available from http://www.bic.kyoto-u.ac.jp/pathway/timhancock.
Collapse
Affiliation(s)
- Timothy Hancock
- Bioinformatics Center, Institute for Chemical Research, Kyoto University, Japan.
| | | | | |
Collapse
|
35
|
Yang T, Xu X, Kernan T, Wu V, Colecraft HM. Rem, a member of the RGK GTPases, inhibits recombinant CaV1.2 channels using multiple mechanisms that require distinct conformations of the GTPase. J Physiol 2010; 588:1665-81. [PMID: 20308247 DOI: 10.1113/jphysiol.2010.187203] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Rad/Rem/Gem/Kir (RGK) GTPases potently inhibit Ca(V)1 and Ca(V)2 (Ca(V)1-2) channels, a paradigm of ion channel regulation by monomeric G-proteins with significant physiological ramifications and potential biotechnology applications. The mechanism(s) underlying how RGK proteins inhibit I(Ca) is unknown, and it is unclear how key structural and regulatory properties of these GTPases (such as the role of GTP binding to the nucleotide binding domain (NBD), and the C-terminus which contains a membrane-targeting motif) feature in this effect. Here, we show that Rem inhibits Ca(V)1.2 channels by three independent mechanisms that rely on distinct configurations of the GTPase: (1) a reduction in surface density of channels is accomplished by enhancing dynamin-dependent endocytosis, (2) a diminution of channel open probability (P(o)) that occurs without impacting on voltage sensor movement, and (3) an immobilization of Ca(V) channel voltage sensors. The presence of both the Rem NBD and C-terminus (whether membrane-targeted or not) in one molecule is sufficient to reconstitute all three mechanisms. However, membrane localization of the NBD by a generic membrane-targeting module reconstitutes only the decreased P(o) function (mechanism 2). A point mutation that prevents GTP binding to the NBD selectively eliminates the capacity to immobilize voltage sensors (mechanism 3). The results reveal an uncommon multiplicity in the mechanisms Rem uses to inhibit I(Ca), predict new physiological dimensions of the RGK GTPase-Ca(V) channel crosstalk, and suggest original approaches for developing novel Ca(V) channel blockers.
Collapse
Affiliation(s)
- Tingting Yang
- Department of Physiology and Cellular Biophysics, Columbia University, College of Physicians and Surgeons, New York, NY 10032, USA
| | | | | | | | | |
Collapse
|
36
|
Abstract
Glucose-stimulated insulin secretion from the islet beta-cell involves a sequence of metabolic events and an interplay between a wide range of signaling pathways leading to the generation of second messengers (e.g., cyclic nucleotides, adenine and guanine nucleotides, soluble lipid messengers) and mobilization of calcium ions. Consequent to the generation of necessary signals, the insulin-laden secretory granules are transported from distal sites to the plasma membrane for fusion and release of their cargo into the circulation. The secretory granule transport underlies precise changes in cytoskeletal architecture involving a well-coordinated cross-talk between various signaling proteins, including small molecular mass GTP-binding proteins (G proteins) and their respective effector proteins. The purpose of this article is to provide an overview of current understanding of the identity of small G proteins (e.g., Cdc42, Rac1, and ARF-6) and their corresponding regulatory factors (e.g., GDP/GTP-exchange factors, GDP-dissociation inhibitors) in the pancreatic beta-cell. Plausible mechanisms underlying regulation of these signaling proteins by insulin secretagogues are also discussed. In addition to their positive modulatory roles, certain small G proteins also contribute to the metabolic dysfunction and demise of the islet beta-cell seen in in vitro and in vivo models of impaired insulin secretion and diabetes. Emerging evidence also suggests significant insulin secretory abnormalities in small G protein knockout animals, further emphasizing vital roles for these proteins in normal health and function of the islet beta-cell. Potential significance of these experimental observations from multiple laboratories and possible avenues for future research in this area of islet research are highlighted.
Collapse
Affiliation(s)
- Anjaneyulu Kowluru
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48202-3489, USA.
| |
Collapse
|
37
|
Treharne KJ, Giles Best O, Mehta A. Transglutaminase 2 and nucleoside diphosphate kinase activity are correlated in epithelial membranes and are abnormal in cystic fibrosis. FEBS Lett 2009; 583:2789-92. [PMID: 19619546 PMCID: PMC2741570 DOI: 10.1016/j.febslet.2009.07.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2009] [Revised: 06/16/2009] [Accepted: 07/14/2009] [Indexed: 11/28/2022]
Abstract
Tissue transglutaminase (tgase2) is a multifunctional enzyme that crosslinks proteins but also acts as a G-protein, differential functions regulated by calcium and GTP. In the epithelial cell membrane, we show that manipulation of tgase2 function by monodansylcadaverine or retinoic acid (RA) alters the activity of a membrane-bound protein kinase, nucleoside diphosphate kinase (NDPK, nm23-H1/H2) that is known to control G-protein function. We find that NDPK function is abnormally low in cystic fibrosis but can be restored by RA treatment in vitro. Our data suggest that tgase2 is overexpressed in cystic fibrosis and affects NDPK function.
Collapse
Affiliation(s)
- Kate J Treharne
- Centre for Cardiovascular and Lung Biology, Division of Medical Sciences, Ninewells Hospital and Medical School, University of Dundee, United Kingdom
| | | | | |
Collapse
|
38
|
Kaul R, Murakami M, Lan K, Choudhuri T, Robertson ES. EBNA3C can modulate the activities of the transcription factor Necdin in association with metastasis suppressor protein Nm23-H1. J Virol 2009; 83:4871-83. [PMID: 19116252 PMCID: PMC2682100 DOI: 10.1128/jvi.02286-08] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Previous studies have demonstrated the interaction between the Epstein-Barr virus (EBV) nuclear antigen 3C (EBNA3C) and the metastatic suppressor Nm23-H1 both in vitro and in vivo (C. Subramanian, M. A. Cotter II, and E. S. Robertson, Nat. Med. 7:350-355, 2001). Importantly EBNA3C can reverse the ability of Nm23-H1 to suppress migration of human cells in vitro. EBNA3C contributes to EBV-associated human cancers by regulating transcription of a number of cellular and viral promoters as well as targeting and altering the transcription activities of the metastasis suppressor Nm23-H1. Furthermore, Necdin is a cellular protein which is highly induced in terminally differentiated cells; it contributes to the regulation of cell growth and is also known to interact with viral oncoproteins. In this report, we show that Nm23-H1 and EBNA3C can modulate the biological functions of Necdin in the context of EBV infection and transformation. The levels of Necdin were consistently lower in EBV-positive cells, and EBNA3C could change the subcellular localization of Necdin as well as rescue cells from the antiangiogenic and antiproliferative effects mediated by Necdin. We also show that Necdin directly interacts with Nm23-H1, resulting in modulation of the biochemical function of Nm23-H1 as well as the biological function of Necdin. Both EBNA3C and Nm23-H1 were able to rescue not only Necdin-mediated transcriptional repression of the downstream vascular endothelial growth factor promoter but also Necdin-mediated growth suppression and antiangiogenic effects on cancer cells. The majority of this response was mediated through amino acid residues 191 to 222 of Necdin, which are also known to be important for nuclear matrix targeting. These studies suggest a role for Necdin in the regulation of downstream cellular targets in a hypoxic environment in virus-associated human cancers.
Collapse
Affiliation(s)
- Rajeev Kaul
- Department of Microbiology and Tumor Virology Program, Abramson Comprehensive Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|
39
|
Galasso A, Zollo M. The Nm23-H1-h-Prune complex in cellular physiology: a 'tip of the iceberg' protein network perspective. Mol Cell Biochem 2009; 329:149-59. [PMID: 19390954 DOI: 10.1007/s11010-009-0115-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2009] [Accepted: 04/02/2009] [Indexed: 02/08/2023]
Abstract
Nm23-H1 (also known as NDPKA) and h-Prune form a protein complex that is part of a little-understood protein network. Modifications of this complex correlate with cancer status. Here, we focus on the role of the Nm23-H1-h-Prune complex in cellular physiology, through an analysis of the balance between the 'bound' and 'non-bound' states of Nm23-H1 and h-Prune, whereby we speculate on the 'read-out' during cell homeostasis under non-balanced conditions. We have analysed the biochemical activities of both Nm23-H1 and h-Prune alone and in combination, focussing on the anti-metastatic activity of Nm23-H1. We have then investigated the cellular mechanisms responsible for the formation of the Nm23-H1-h-Prune complex. To evaluate the importance of the equilibrium between the formation of the Nm23-H1-h-Prune complex and the 'free' levels of Nm23-H1 and h-Prune, we propose a model based on a pro-cancer condition where this equilibrium is negatively affected.
Collapse
Affiliation(s)
- Alessia Galasso
- Centro di Ingegneria Genetica e Biotecnologia Avanzate (CEINGE), Via Comunale Margherita 482, 80145 Naples, Italy
| | | |
Collapse
|
40
|
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
|
41
|
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: 160] [Impact Index Per Article: 10.0] [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
|
42
|
Kim HD, Youn B, Kim TS, Kim SH, Shin HS, Kim J. Regulators affecting the metastasis suppressor activity of Nm23-H1. Mol Cell Biochem 2009. [PMID: 19377884 DOI: 10.1007/s11010-] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Nm23-H1 encodes nucleoside diphosphate kinase A (NDPK-A) and is known to have a metastasis suppressive activity in many tumor cells. However, it has many other functions as well. Recent studies have shown that the interacting proteins with Nm23-H1 which mediate the cell proliferation, may act as modulators of the metastasis suppressor activity. The interacting proteins with Nm23-H1 can be classified into 3 groups. The first group of proteins can be classified as upstream kinases of Nm23-H1 such as CKI and Aurora-A/STK15. The second group of proteins acts as downstream effectors for the regulation of specific gene transcriptions, GTP-binding protein functions, and signal transduction in Erk signal cascade. The third group of proteins can be classified as bi-directionally influencing binding partners of Nm23-H1. As a result, the interactions with Nm23-H1 and binding partners have implications in the biochemical characterization involved in metastasis and tumorigenesis.
Collapse
Affiliation(s)
- Hag Dong Kim
- School of Life Sciences & Biotechnology, and BioInstitute, Korea University, Seoul 136-713, Korea
| | | | | | | | | | | |
Collapse
|
43
|
Kim HD, Youn B, Kim TS, Kim SH, Shin HS, Kim J. Regulators affecting the metastasis suppressor activity of Nm23-H1. Mol Cell Biochem 2009; 329:167-73. [PMID: 19377884 DOI: 10.1007/s11010-009-0109-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2009] [Accepted: 04/02/2009] [Indexed: 10/20/2022]
Abstract
Nm23-H1 encodes nucleoside diphosphate kinase A (NDPK-A) and is known to have a metastasis suppressive activity in many tumor cells. However, it has many other functions as well. Recent studies have shown that the interacting proteins with Nm23-H1 which mediate the cell proliferation, may act as modulators of the metastasis suppressor activity. The interacting proteins with Nm23-H1 can be classified into 3 groups. The first group of proteins can be classified as upstream kinases of Nm23-H1 such as CKI and Aurora-A/STK15. The second group of proteins acts as downstream effectors for the regulation of specific gene transcriptions, GTP-binding protein functions, and signal transduction in Erk signal cascade. The third group of proteins can be classified as bi-directionally influencing binding partners of Nm23-H1. As a result, the interactions with Nm23-H1 and binding partners have implications in the biochemical characterization involved in metastasis and tumorigenesis.
Collapse
Affiliation(s)
- Hag Dong Kim
- School of Life Sciences & Biotechnology, and BioInstitute, Korea University, Seoul 136-713, Korea
| | | | | | | | | | | |
Collapse
|
44
|
Jin L, Liu G, Zhang CH, Lu CH, Xiong S, Zhang MY, Liu QY, Ge F, He QY, Kitazato K, Kobayashi N, Wang YF. Nm23-H1 regulates the proliferation and differentiation of the human chronic myeloid leukemia K562 cell line: a functional proteomics study. Life Sci 2009; 84:458-67. [PMID: 19302816 DOI: 10.1016/j.lfs.2009.01.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2008] [Revised: 01/05/2009] [Accepted: 01/23/2009] [Indexed: 11/18/2022]
Abstract
AIMS Nm23-H1 is a suppressor of metastasis that has been implicated in the regulation of proliferation and differentiation of hematopoietic cells, although specific mechanisms for Nm23-H1 have not been well-characterized. Our study is designed to further elucidate the role of Nm23-H1 in the human chronic myeloid leukemia K562 cell line. MAIN METHODS In this study we generated and selected two cell clone pools of human chronic myeloid leukemia K562 cells with up-regulated and down-regulated Nm23-H1 expression. KEY FINDINGS Our data show that knockdown of Nm23-H1 decreased proliferation and increased the percentage of cells arrested in the G0/G1 phase of the cell cycle. Correspondingly, K562 cells overexpressing Nm23-H1 were more proliferative. After treatment of these two cell types with phorbol 12-myristate 13-acetate (PMA) for 48 h, cells with reduced Nm23-H1 expression had a higher percentage of 8N ploidy and higher expression of CD41 than K562 cells overexpressing Nm23-H1. A functional proteomics analysis identified ten proteins, including ANP32A, Cdc42GAP, and the isoform 2 of SET, whose expression levels were significantly altered by down-regulation of Nm23-H1. In addition, cells with decreased levels of Nm23-H1 had significantly reduced expression of Cdc42 independent of treatment with PMA. The interaction of the endogenous Nm23-H1 and Cdc42 proteins has been further validated by reciprocal immunoprecipitations. SIGNIFICANCE We provide data that complement functional studies of Nm23-H1 in regulating hematopoietic cells, and address action mechanisms of Nm23-H1 that have not previously been reported.
Collapse
Affiliation(s)
- Lin Jin
- The Institute of Pharmacology Science, Jinan University, Guangzhou, 510632 China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Youn B, Kim HD, Kim J. Nm23-H1/nucleoside diphosphate kinase as a key molecule in breast tumor angiogenesis. Expert Opin Ther Targets 2009; 12:1419-30. [PMID: 18851697 DOI: 10.1517/14728222.12.11.1419] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Neo-angiogenesis seems to be a critical feature of breast tumor growth, migration and metastasis. Inhibition of angiogenesis may provide information regarding treatment. Since angiogenesis is the result of complex processes, controlled by several angiogenic (pro- and/or -anti) factors and their receptors, multiple ways to prevent or retrogress tumor-induced angiogenesis have been proposed. The clinically significant activity of bevacizumab and other antiangiogenic treatments have attracted a great deal of interest. OBJECTIVE/METHODS We discuss biological aspects of breast cancer angiogenesis and nucleoside diphosphate kinase (NDPK) as a key molecule in this process. RESULTS/CONCLUSIONS In clinical and experimental trials, it was reported that NDPK is inversely related to breast cancer metastasis and angiogenesis. To inhibit the metastatic potential of cancer cells, Nm23-H1/NDP kinase appears to interact with many proteins involved in cellular signal transduction in angiogenesis and tumorigenesis, and therefore reduces the activation of the extracellular signal-regulated kinase (ERK)/MAPK in response to those signals.
Collapse
Affiliation(s)
- BuHyun Youn
- Division of Biological Sciences, Pusan National University, College of Natural Sciences, Busan, 609-735, Korea
| | | | | |
Collapse
|
46
|
Shen Y, Han YJ, Kim JI, Song PS. Arabidopsis nucleoside diphosphate kinase-2 as a plant GTPase activating protein. BMB Rep 2008; 41:645-50. [PMID: 18823588 DOI: 10.5483/bmbrep.2008.41.9.645] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Nucleoside diphosphate kinase (NDPK) is involved in multiple signaling pathways in mammalian systems, including G-protein signaling. Arabidopsis NDPK2, like its mammalian counterparts, is multifunctional despite its initial discovery phytochrome-interacting protein. This similarity raises the possibility that NDPK2 may play a role in G-protein signaling in plants. In the present study, we explore the potential relationship between NDPK2 and the small G proteins, Pra2 and Pra3, as well as the heterotrimeric G protein, GPA1. We report a physical interaction between NDPK2 and these small G proteins, and demonstrate that NDPK2 can stimulate their GTPase activities. Our results suggest that NDPK2 acts as a GTPase-activating protein for small G proteins in plants. We propose that NDPK2 might be a missing link between the phytochromemediated light signaling and G protein-mediated signaling.
Collapse
Affiliation(s)
- Yu Shen
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | | | | | | |
Collapse
|
47
|
Shi XZ, Zhao XF, Wang JX. Molecular cloning and analysis of function of nucleoside diphosphate kinase (NDPK) from the scallop Chlamys farreri. BIOCHEMISTRY (MOSCOW) 2008; 73:686-92. [PMID: 18620535 DOI: 10.1134/s0006297908060096] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Nucleoside diphosphate kinase (NDPK) is a key metabolic enzyme that catalyzes the synthesis of non-adenine nucleoside triphosphate (NTP) by transferring the terminal phosphate between nucleoside diphosphate (NDP) and NTP. NDPK regulates a variety of eukaryotic cellular activities including cell proliferation, development, and differentiation. The ndpk cDNA was cloned from the hemocytes of the scallop Chlamys farreri and designated Cf-ndpk. The full-length sequence of Cf-ndpk consists of 715 bp encoding a polypeptide of 153 amino acids with a calculated molecular mass of 16927.52 daltons and pI of 7.64. The mRNA expression and distribution of Cf-ndpk in both bacterially challenged and unchallenged scallops were studied by Northern blotting and in situ hybridization. The results showed that Cf-ndpk transcripts were present in hemocytes, gill, adductor muscle, mantle, digestive gland, foot, and gonad, and the expression level increased in hemocytes after bacterial challenge. Recombinant Cf-NDPK expressed in Escherichia coli could transfer the terminal phosphate between UDP and ATP. The Cf-NDPK protein was present in all tested tissues including foot, adductor muscle, digestive gland, gonad, mantle, gill, and hemolymph. It was up-regulated in hemolymph after bacterial challenge. Taken together, these results suggest that NDPK may play roles in the innate immune response of scallop.
Collapse
Affiliation(s)
- Xiu-Zhen Shi
- School of Life Sciences, Shandong University, Jinan, Shandong 250100, China.
| | | | | |
Collapse
|
48
|
Fischbach MA, Settleman J. Regulation of the nucleotide state of oncogenic ras proteins by nucleoside diphosphate kinase. Methods Enzymol 2008; 407:33-45. [PMID: 16757312 DOI: 10.1016/s0076-6879(05)07004-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
Oncogenic forms of the Ras GTPase exhibit defective GTP hydrolase activity and are insensitive to the stimulatory activity of GTPase activating proteins. It has been suggested that a potential therapeutic strategy to inactivate such mutant forms of Ras could involve small molecules that restore GTP hydrolase activity to mutant Ras proteins; however, thus far, such molecules have not been developed. While characterizing the biochemical properties of several commonly detected K-Ras mutants, we made the unexpected observation that an activity in crude bacterial cell extracts was capable of stimulating the conversion of the oncogenic K-RasG13D mutant from a GTP-bound, active form to a GDP-bound, inactive form. The activity was purified, and the protein, nucleoside diphosphate kinase (NDK), was identified as being responsible for the Ras regulating activity. NDK is closely related to the human metastasis suppressor, NM23, which has previously been implicated in regulating the nucleotide state of small GTPases of the Ras family. Although the physiological relevance of such regulation has been controversial, our biochemical findings in in vitro assays indicate that it may be feasible to develop a therapeutic strategy to achieve the selective biochemical inactivation of oncogenic Ras proteins.
Collapse
Affiliation(s)
- Michael A Fischbach
- General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts, USA
| | | |
Collapse
|
49
|
Kowluru A. Emerging roles for protein histidine phosphorylation in cellular signal transduction: lessons from the islet beta-cell. J Cell Mol Med 2008; 12:1885-908. [PMID: 18400053 PMCID: PMC4506158 DOI: 10.1111/j.1582-4934.2008.00330.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Protein phosphorylation represents one of the key regulatory events in physiological insulin secretion from the islet β-cell. In this context, several classes of protein kinases (e.g. calcium-, cyclic nucleotide- and phospholipid-dependent protein kinases and tyrosine kinases) have been characterized in the β-cell. The majority of phosphorylated amino acids identified include phosphoserine, phosphothreonine and phosphotyrosine. Protein histidine phosphorylation has been implicated in the prokaryotic and eukaryotic cellular signal transduction. Most notably, phoshohistidine accounts for 6% of total protein phosphorylation in eukaryotes, which makes it nearly 100-fold more abundant than phosphotyrosine, but less abundant than phosphoserine and phosphothreonine. However, very little is known about the number of proteins with phosphohistidines, since they are highly labile and are rapidly lost during phosphoamino acid identification under standard experimental conditions. The overall objectives of this review are to: (i) summarize the existing evidence indicating the subcellular distribution and characterization of various histidine kinases in the islet β-cell, (ii) describe evidence for functional regulation of these kinases by agonists of insulin secretion, (iii) present a working model to implicate novel regulatory roles for histidine kinases in the receptor-independent activation, by glucose, of G-proteins endogenous to the β-cell, (iv) summarize evidence supporting the localization of protein histidine phosphatases in the islet β-cell and (v) highlight experimental evidence suggesting potential defects in the histidine kinase signalling cascade in islets derived from the Goto-Kakizaki (GK) rat, a model for type 2 diabetes. Potential avenues for future research to further decipher regulatory roles for protein histidine phosphorylation in physiological insulin secretion are also discussed.
Collapse
Affiliation(s)
- Anjaneyulu Kowluru
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA.
| |
Collapse
|
50
|
Correll RN, Pang C, Niedowicz DM, Finlin BS, Andres DA. The RGK family of GTP-binding proteins: regulators of voltage-dependent calcium channels and cytoskeleton remodeling. Cell Signal 2008; 20:292-300. [PMID: 18042346 PMCID: PMC2254326 DOI: 10.1016/j.cellsig.2007.10.028] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2007] [Accepted: 10/30/2007] [Indexed: 02/05/2023]
Abstract
RGK proteins constitute a novel subfamily of small Ras-related proteins that function as potent inhibitors of voltage-dependent (VDCC) Ca(2+) channels and regulators of actin cytoskeletal dynamics. Within the larger Ras superfamily, RGK proteins have distinct regulatory and structural characteristics, including nonconservative amino acid substitutions within regions known to participate in nucleotide binding and hydrolysis and a C-terminal extension that contains conserved regulatory sites which control both subcellular localization and function. RGK GTPases interact with the VDCC beta-subunit (Ca(V)beta) and inhibit Rho/Rho kinase signaling to regulate VDCC activity and the cytoskeleton respectively. Binding of both calmodulin and 14-3-3 to RGK proteins, and regulation by phosphorylation controls cellular trafficking and the downstream signaling of RGK proteins, suggesting that a complex interplay between interacting protein factors and trafficking contribute to their regulation.
Collapse
Affiliation(s)
- Robert N Correll
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY 40536, United States
| | | | | | | | | |
Collapse
|