1
|
Xu X, Jin T. Ras inhibitors gate chemoattractant concentration range for chemotaxis through controlling GPCR-mediated adaptation and cell sensitivity. Front Immunol 2022; 13:1020117. [PMID: 36341344 PMCID: PMC9630474 DOI: 10.3389/fimmu.2022.1020117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 10/03/2022] [Indexed: 11/13/2022] Open
Abstract
Chemotaxis plays an essential role in recruitment of leukocytes to sites of inflammation. Eukaryotic cells sense chemoattractant with G protein-coupled receptors (GPCRs) and chemotax toward gradients with an enormous concentration range through adaptation. Cells in adaptation no longer respond to the present stimulus but remain sensitive to stronger stimuli. Thus, adaptation provides a fundamental strategy for eukaryotic cells to chemotax through a gradient. Ras activation is the first step in the chemosensing GPCR signaling pathways that displays a transient activation behavior in both model organism Dictyostelium discoideum and mammalian neutrophils. Recently, it has been revealed that C2GAP1 and CAPRI control the GPCR-mediated adaptation in D. discoideum and human neutrophils, respectively. More importantly, both Ras inhibitors regulate the sensitivity of the cells. These findings suggest an evolutionarily conserved molecular mechanism by which eukaryotic cells gate concentration range of chemoattractants for chemotaxis.
Collapse
|
2
|
Ras inhibitor CAPRI enables neutrophil-like cells to chemotax through a higher-concentration range of gradients. Proc Natl Acad Sci U S A 2021; 118:2002162118. [PMID: 34675073 DOI: 10.1073/pnas.2002162118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2021] [Indexed: 01/21/2023] Open
Abstract
Neutrophils sense and migrate through an enormous range of chemoattractant gradients through adaptation. Here, we reveal that in human neutrophils, calcium-promoted Ras inactivator (CAPRI) locally controls the GPCR-stimulated Ras adaptation. Human neutrophils lacking CAPRI (caprikd ) exhibit chemoattractant-induced, nonadaptive Ras activation; significantly increased phosphorylation of AKT, GSK-3α/3β, and cofilin; and excessive actin polymerization. caprikd cells display defective chemotaxis in response to high-concentration gradients but exhibit improved chemotaxis in low- or subsensitive-concentration gradients of various chemoattractants, as a result of their enhanced sensitivity. Taken together, our data reveal that CAPRI controls GPCR activation-mediated Ras adaptation and lowers the sensitivity of human neutrophils so that they are able to chemotax through a higher-concentration range of chemoattractant gradients.
Collapse
|
3
|
Kolakowska J, Drzewiecka EM, Kozlowska W, Zmijewska A, Souchelnytskyi S, Franczak A. Proteomic profile alterations in porcine conceptuses during early stages of development. Reprod Biol 2021; 21:100481. [PMID: 33529998 DOI: 10.1016/j.repbio.2021.100481] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 12/02/2020] [Accepted: 01/15/2021] [Indexed: 11/25/2022]
Abstract
The dynamic embryo development during the early stages of gestation requires precise molecular changes, including proteomic ones. We aimed to find unique proteins for porcine conceptuses specifically during the peri-implantation period, i.e. on days 15-16 of pregnancy. The proteomic profile of these conceptuses was compared with conceptuses at an earlier stage of the development, i.e. collected during maternal recognition of pregnancy on days 12-13 of pregnancy. The 2DE, gel image analysis, and MALDI TOF mass spectrometry were used 500 protein spots were annotated as common to conceptuses harvested during both studied periods. Proteomic profile of the conceptuses collected during the peri-implantation period contains 24 unique proteins. Identified unique for the peri-implantation period proteins are involved in adhesion processes, cadherin, and actin-binding, and actin filament organization, extracellular matrix organization, and cytoskeleton organization. Systemic analysis of identified proteins confirmed their involvement in cell adhesion and cytoskeletal organization as being two major affected functions. The unique proteins might be recognized as factors conditioning the proper peri-implantation embryo development and gaining competences for implantation. In further studies, BRCA1 might be considered as a candidate for a potential marker of embryonic competences for implantation in pigs.
Collapse
Affiliation(s)
- Justyna Kolakowska
- Department of Anatomy and Animal Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowski 1A, 10-719, Olsztyn, Poland
| | - Ewa Monika Drzewiecka
- Department of Anatomy and Animal Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowski 1A, 10-719, Olsztyn, Poland
| | - Wiktoria Kozlowska
- Department of Anatomy and Animal Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowski 1A, 10-719, Olsztyn, Poland
| | - Agata Zmijewska
- Department of Anatomy and Animal Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowski 1A, 10-719, Olsztyn, Poland
| | - Serhiy Souchelnytskyi
- Neurocentrum, Karolinska University Hospital, Neurologmottagningen, 171 76, Solna, Stockholm, Sweden; Oranta Cancer Diagnostics AB, Norrens väg 73, 752 63, Uppsala, Sweden; College of Medicine, QU Health, Qatar University, Building H12, PO Box 2713, Doha, Qatar
| | - Anita Franczak
- Department of Anatomy and Animal Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowski 1A, 10-719, Olsztyn, Poland.
| |
Collapse
|
4
|
Ayad O, Al Sayed ZR, Sebille S, Magaud C, Chapotte-Baldacci CA, Jayle C, Faivre JF, Gaborit N, Chatelier A, Bois P. In vitro differentiation of W8B2 + human cardiac stem cells: gene expression of ionic channels and spontaneous calcium activity. Cell Mol Biol Lett 2020; 25:50. [PMID: 33292162 PMCID: PMC7646077 DOI: 10.1186/s11658-020-00242-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 10/29/2020] [Indexed: 11/18/2022] Open
Abstract
Background Human cardiac stem cells expressing the W8B2 marker (W8B2+ CSCs) were recently identified and proposed as a new model of multipotent CSCs capable of differentiating into smooth muscle cells, endothelial cells and immature myocytes. Nevertheless, no characterization of ion channel or calcium activity during the differentiation of these stem cells has been reported. Methods The objectives of this study were thus to analyze (using the TaqMan Low-Density Array technique) the gene profile of W8B2+ CSCs pertaining to the regulation of ion channels, transporters and other players involved in the calcium homeostasis of these cells. We also analyzed spontaneous calcium activity (via the GCaMP calcium probe) during the in vitro differentiation of W8B2+ CSCs into cardiac myocytes. Results Our results show an entirely different electrophysiological genomic profile between W8B2+ CSCs before and after differentiation. Some specific nodal genes, such as Tbx3, HCN, ICaT, L, KV, and NCX, are overexpressed after this differentiation. In addition, we reveal spontaneous calcium activity or a calcium clock whose kinetics change during the differentiation process. A pharmacological study carried out on differentiated W8B2+ CSCs showed that the NCX exchanger and IP3 stores play a fundamental role in the generation of these calcium oscillations. Conclusions Taken together, the present results provide important information on ion channel expression and intrinsic calcium dynamics during the differentiation process of stem cells expressing the W8B2 marker.
Collapse
Affiliation(s)
- Oualid Ayad
- University of Poitiers Signalisation et Transports Ioniques Membranaires, EA7349, Poitiers Cedex 09, France
| | - Zeina R Al Sayed
- CNRS, INSERM, l'institut du thorax, Université de Nantes, 44000, Nantes, France
| | - Stéphane Sebille
- University of Poitiers Signalisation et Transports Ioniques Membranaires, EA7349, Poitiers Cedex 09, France
| | - Christophe Magaud
- University of Poitiers Signalisation et Transports Ioniques Membranaires, EA7349, Poitiers Cedex 09, France
| | | | - Christophe Jayle
- CHU of Poitiers chirurgie cardiaque et thoracique, , Poitiers Cedex 09, France
| | - Jean-François Faivre
- University of Poitiers Signalisation et Transports Ioniques Membranaires, EA7349, Poitiers Cedex 09, France
| | - Nathalie Gaborit
- CNRS, INSERM, l'institut du thorax, Université de Nantes, 44000, Nantes, France
| | - Aurélien Chatelier
- University of Poitiers Signalisation et Transports Ioniques Membranaires, EA7349, Poitiers Cedex 09, France
| | - Patrick Bois
- University of Poitiers Signalisation et Transports Ioniques Membranaires, EA7349, Poitiers Cedex 09, France.
| |
Collapse
|
5
|
Bellazzo A, Collavin L. Cutting the Brakes on Ras-Cytoplasmic GAPs as Targets of Inactivation in Cancer. Cancers (Basel) 2020; 12:cancers12103066. [PMID: 33096593 PMCID: PMC7588890 DOI: 10.3390/cancers12103066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/11/2020] [Accepted: 10/15/2020] [Indexed: 12/16/2022] Open
Abstract
Simple Summary GTPase-Activating Proteins (RasGAPs) are a group of structurally related proteins with a fundamental role in controlling the activity of Ras in normal and cancer cells. In particular, loss of function of RasGAPs may contribute to aberrant Ras activation in cancer. Here we review the multiple molecular mechanisms and factors that are involved in downregulating RasGAPs expression and functions in cancer. Additionally, we discuss how extracellular stimuli from the tumor microenvironment can control RasGAPs expression and activity in cancer cells and stromal cells, indirectly affecting Ras activation, with implications for cancer development and progression. Abstract The Ras pathway is frequently deregulated in cancer, actively contributing to tumor development and progression. Oncogenic activation of the Ras pathway is commonly due to point mutation of one of the three Ras genes, which occurs in almost one third of human cancers. In the absence of Ras mutation, the pathway is frequently activated by alternative means, including the loss of function of Ras inhibitors. Among Ras inhibitors, the GTPase-Activating Proteins (RasGAPs) are major players, given their ability to modulate multiple cancer-related pathways. In fact, most RasGAPs also have a multi-domain structure that allows them to act as scaffold or adaptor proteins, affecting additional oncogenic cascades. In cancer cells, various mechanisms can cause the loss of function of Ras inhibitors; here, we review the available evidence of RasGAP inactivation in cancer, with a specific focus on the mechanisms. We also consider extracellular inputs that can affect RasGAP levels and functions, implicating that specific conditions in the tumor microenvironment can foster or counteract Ras signaling through negative or positive modulation of RasGAPs. A better understanding of these conditions might have relevant clinical repercussions, since treatments to restore or enhance the function of RasGAPs in cancer would help circumvent the intrinsic difficulty of directly targeting the Ras protein.
Collapse
|
6
|
Yang B, Li R, Liu PN, Geng X, Mooney BP, Chen C, Cheng J, Fritsche KL, Beversdorf DQ, Lee JC, Sun GY, Greenlief CM. Quantitative Proteomics Reveals Docosahexaenoic Acid-Mediated Neuroprotective Effects in Lipopolysaccharide-Stimulated Microglial Cells. J Proteome Res 2020; 19:2236-2246. [PMID: 32302149 PMCID: PMC7282485 DOI: 10.1021/acs.jproteome.9b00792] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
![]()
The high levels of docosahexaenoic
acid (DHA) in cell membranes
within the brain have led to a number of studies exploring its function.
These studies have shown that DHA can reduce inflammatory responses
in microglial cells. However, the method of action is poorly understood.
Here, we report the effects of DHA on microglial cells stimulated
with lipopolysaccharides (LPSs). Data were acquired using the parallel
accumulation serial fragmentation method in a hybrid trapped ion mobility-quadrupole
time-of-flight mass spectrometer. Over 2800 proteins are identified
using label-free quantitative proteomics. Cells exposed to LPSs and/or
DHA resulted in changes in cell morphology and expression of 49 proteins
with differential abundance (greater than 1.5-fold change). The data
provide details about pathways that are influenced in this system
including the nuclear factor κ-light-chain-enhancer of the activated
B cells (NF-κB) pathway. Western blots and enzyme-linked immunosorbent
assay studies are used to help confirm the proteomic results. The
MS data are available at ProteomeXchange.
Collapse
Affiliation(s)
- Bo Yang
- Department of Chemistry, University of Missouri, Columbia 65211, Missouri, United States.,Charles W. Gehrke Proteomics Center, University of Missouri, Columbia 65211, Missouri, United States
| | - Runting Li
- Biochemistry Department, University of Missouri, Columbia 65211, Missouri, United States
| | - Pei N Liu
- Charles W. Gehrke Proteomics Center, University of Missouri, Columbia 65211, Missouri, United States
| | - Xue Geng
- Department of Bioengineering, University of Illinois at Chicago, Chicago 60612, Illinois, United States
| | - Brian P Mooney
- Biochemistry Department, University of Missouri, Columbia 65211, Missouri, United States.,Charles W. Gehrke Proteomics Center, University of Missouri, Columbia 65211, Missouri, United States
| | - Chen Chen
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia 65211, Missouri, United States
| | - Jianlin Cheng
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia 65211, Missouri, United States
| | - Kevin L Fritsche
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia 65211, Missouri, United States
| | - David Q Beversdorf
- Departments of Radiology, Neurology and Psychological Sciences, and the Thompson Center, University of Missouri, Columbia 65211, Missouri, United States
| | - James C Lee
- Department of Bioengineering, University of Illinois at Chicago, Chicago 60612, Illinois, United States
| | - Grace Y Sun
- Biochemistry Department, University of Missouri, Columbia 65211, Missouri, United States
| | - C Michael Greenlief
- Department of Chemistry, University of Missouri, Columbia 65211, Missouri, United States.,Charles W. Gehrke Proteomics Center, University of Missouri, Columbia 65211, Missouri, United States
| |
Collapse
|
7
|
Thaker YR, Raab M, Strebhardt K, Rudd CE. GTPase-activating protein Rasal1 associates with ZAP-70 of the TCR and negatively regulates T-cell tumor immunity. Nat Commun 2019; 10:4804. [PMID: 31641113 PMCID: PMC6805919 DOI: 10.1038/s41467-019-12544-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Accepted: 09/09/2019] [Indexed: 12/11/2022] Open
Abstract
Immunotherapy involving checkpoint blockades of inhibitory co-receptors is effective in combating cancer. Despite this, the full range of mediators that inhibit T-cell activation and influence anti-tumor immunity is unclear. Here, we identify the GTPase-activating protein (GAP) Rasal1 as a novel TCR-ZAP-70 binding protein that negatively regulates T-cell activation and tumor immunity. Rasal1 inhibits via two pathways, the binding and inhibition of the kinase domain of ZAP-70, and GAP inhibition of the p21ras-ERK pathway. It is expressed in activated CD4 + and CD8 + T-cells, and inhibits CD4 + T-cell responses to antigenic peptides presented by dendritic cells as well as CD4 + T-cell responses to peptide antigens in vivo. Furthermore, siRNA reduction of Rasal1 expression in T-cells shrinks B16 melanoma and EL-4 lymphoma tumors, concurrent with an increase in CD8 + tumor-infiltrating T-cells expressing granzyme B and interferon γ-1. Our findings identify ZAP-70-associated Rasal1 as a new negative regulator of T-cell activation and tumor immunity. Activation of T cells in the tumor microenvironment can be inhibited through a variety of mechanisms. Here, the authors show that Rasal1, a GTPase-activating protein, binds and inhibits signaling downstream of the T Cell Receptor complex and that consistently, its reduced expression enhances anti-tumor T-cell responses in two syngeneic cancer mouse models.
Collapse
Affiliation(s)
- Youg Raj Thaker
- Cell Signalling Section, Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK.,School of Biological Science, Protein Structure and Disease Mechanisms, University of Essex, Wivenhoe Park, Colchester, CO4 3SQ, UK
| | - Monika Raab
- Department of Obstetrics and Gynaecology, School of Medicine, J.W. Goethe-University, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Klaus Strebhardt
- Department of Obstetrics and Gynaecology, School of Medicine, J.W. Goethe-University, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Christopher E Rudd
- Cell Signalling Section, Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK. .,Département de Immunologie-Oncologie, Centre de Recherche Hôpital Maisonneuve-Rosemont, Montreal, QC, H1T 2M4, Canada. .,Département de Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada.
| |
Collapse
|
8
|
Li R, Grimm SA, Mav D, Gu H, Djukovic D, Shah R, Merrick BA, Raftery D, Wade PA. Transcriptome and DNA Methylome Analysis in a Mouse Model of Diet-Induced Obesity Predicts Increased Risk of Colorectal Cancer. Cell Rep 2019; 22:624-637. [PMID: 29346762 PMCID: PMC5793878 DOI: 10.1016/j.celrep.2017.12.071] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 11/16/2017] [Accepted: 12/20/2017] [Indexed: 12/30/2022] Open
Abstract
Colorectal cancer (CRC) tends to occur at older age; however, CRC incidence rates have been rising sharply among young age groups. The increasing prevalence of obesity is recognized as a major risk, yet the mechanistic underpinnings remain poorly understood. Using a diet-induced obesity mouse model, we identified obesity-associated molecular changes in the colonic epithelium of young and aged mice, and we further investigated whether the changes were reversed after weight loss. Transcriptome analysis indicated that obesity-related colonic cellular metabolic switch favoring long-chain fatty acid oxidation happened in young mice, while obesity-associated downregulation of negative feedback regulators of pro-proliferative signaling pathways occurred in older mice. Strikingly, colonic DNA methylome was pre-programmed by obesity at young age, priming for a tumor-prone gene signature after aging. Furthermore, obesity-related changes were substantially preserved after short-term weight loss, but they were largely reversed after long-term weight loss. We provided mechanistic insights into increased CRC risk in obesity.
Collapse
Affiliation(s)
- Ruifang Li
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Sara A Grimm
- Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Deepak Mav
- Sciome, LLC, 2 Davis Drive, Research Triangle Park, NC 27709, USA
| | - Haiwei Gu
- Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine, University of Washington, 850 Republican St., Seattle, WA 98109, USA
| | - Danijel Djukovic
- Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine, University of Washington, 850 Republican St., Seattle, WA 98109, USA
| | - Ruchir Shah
- Sciome, LLC, 2 Davis Drive, Research Triangle Park, NC 27709, USA
| | - B Alex Merrick
- Biomolecular Screening Branch, National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Daniel Raftery
- Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine, University of Washington, 850 Republican St., Seattle, WA 98109, USA; Public Health Sciences Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N., Seattle, WA 98109, USA
| | - Paul A Wade
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
9
|
Response-Related Factors of Bone Marrow-Derived Mesenchymal Stem Cells Transplantation in Patients with Alcoholic Cirrhosis. J Clin Med 2019; 8:jcm8060862. [PMID: 31212896 PMCID: PMC6616969 DOI: 10.3390/jcm8060862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 06/09/2019] [Accepted: 06/14/2019] [Indexed: 12/13/2022] Open
Abstract
Liver cirrhosis leads to hepatic dysfunction and life-threatening conditions. Although the clinical efficacy of autologous bone marrow-derived mesenchymal stem cells (BM-MSC) transplantation in alcoholic cirrhosis (AC) was demonstrated, the relevant mechanism has not been elucidated. We aimed to identify the predictive factors and gene/pathways for responders after autologous BM-MSC transplantation. Fifty-five patients with biopsy-proven AC underwent autologous BM-MSC transplantation. The characteristics of responders who showed improvement in fibrosis score (≥1) after transplantation were compared with those of non-responders. BM-MSCs were analyzed with cDNA microarrays to identify gene/pathways that were differentially expressed in responders. Thirty-three patients (66%) were responders. A high initial Laennec score (p = 0.007, odds ratio 3.73) and performance of BM-MSC transplantation (p = 0.033, odds ratio 5.75) were predictive factors for responders. Three genes (olfactory receptor2L8, microRNA4520-2, and chloride intracellular channel protein3) were upregulated in responders, and CD36 and retinol-binding protein 4 are associated with the biologic processes that are dominant in non-responders. Eleven pathways (inositol phosphate, ATP-binding cassette transporters, protein-kinase signaling, extracellular matrix receptor interaction, endocytosis, phagosome, hematopoietic cell lineage, adipocytokine, peroxisome proliferator-activated receptor, fat digestion/absorption, and insulin resistance) were upregulated in non-responders (p < 0.05). BM-MSC transplantation may be warranted treatment for AC patients with high Laennec scores. Cell-based therapy utilizing response-related genes or pathways can be a treatment candidate.
Collapse
|
10
|
Harrison A, Pentieva K, Ozaki M, McNulty H, Parle-McDermott A. Assessment of candidate folate sensitive-differentially methylated regions in a randomised controlled trial of continued folic acid supplementation during the second and third trimesters of pregnancy. Ann Hum Genet 2018; 83:23-33. [PMID: 30175844 DOI: 10.1111/ahg.12281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 07/06/2018] [Accepted: 07/20/2018] [Indexed: 11/29/2022]
Abstract
SCOPE The aim of this study was to identify if specific regions of the human genome were sensitive to folate status by displaying changes in their DNA methylation patterns in response to continued folic acid supplementation during pregnancy. METHODS AND RESULTS Samples (n = 119) from a previous randomised controlled trial in pregnancy were used to compare the DNA methylation profiles of the same woman pre- versus post-folic acid intervention. Candidate genes were identified from the literature and a pilot genome wide screen of six women (three from each of the folic acid and placebo arms of the trial). We did not observe consistent DNA methylation changes in response to folic acid intervention at any of our candidate genes (RASA4, DHFR, DHFR2, RASSF1A, EIF2C3, ATPF1). We did identify a 40% decrease in DNA methylation at the RASA4 promoter correlating with a 3.5-fold increase in its mRNA abundance in an in vitro cell culture model. CONCLUSION Continued folic acid intervention over a 22-week period did not appear to significantly influence the DNA methylation status of six candidate genes in blood samples of women compared to placebo. However, DNA methylation may play a role in the gene expression control of the RASA4 gene.
Collapse
Affiliation(s)
- Alan Harrison
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Kristina Pentieva
- Nutrition Innovation Centre for Food & Health, Biomedical Sciences Research Institute, Ulster University, Coleraine, United Kingdom
| | - Mari Ozaki
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Helene McNulty
- Nutrition Innovation Centre for Food & Health, Biomedical Sciences Research Institute, Ulster University, Coleraine, United Kingdom
| | | |
Collapse
|
11
|
Jain R, Watson U, Vasudevan L, Saini DK. ERK Activation Pathways Downstream of GPCRs. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 338:79-109. [PMID: 29699693 DOI: 10.1016/bs.ircmb.2018.02.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
GPCRs, the 7-TM receptors, represent a class of cell surface receptors which modulate a variety of physiological responses. The serpentine structure in addition to contributing the diversity of stimuli these receptors can sense also provides flexibility to the extracellular and intracellular regions where other proteins can interact with and can form functionally active multimeric entities. The range in signaling and physiological responses generated by these receptors can be attributed to a large repertoire of the receptor subtypes as well as their differential coupling to various classes of G-protein subunits and other proteins which facilitate multistate activation. A multistate GPCR can engage diverse signaling molecules, thereby modulating not only the canonical cellular responses but also noncanonical responses typically associated with activation of other cascades such as RTK and MAPK/ERK signaling. Given the crucial involvement of MAP kinase/ERK signaling in cell fate determination specially with respect to regulating cell proliferation, cellular apoptosis, and survival, GPCR-mediated cross-activation of MAPK has been explored in various systems and shown to involve functional integration of multiple pathways. This review describes the present knowledge of the different mechanisms of ERK activation downstream of GPCRs and our present understanding of receptor-dependent and -independent MAPK activation cascades.
Collapse
Affiliation(s)
- Ruchi Jain
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
| | - Uchenna Watson
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India; Department of Studies in Zoology, University of Mysore, Manasagangothri, Mysore, India
| | - Lakshmi Vasudevan
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India; L-GEST-Laboratory of Toxicology, Faculty of Pharmaceutical Sciences, Ghent, Belgium
| | - Deepak K Saini
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India; Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India.
| |
Collapse
|
12
|
Li L, Zeng TT, Zhang BZ, Li Y, Zhu YH, Guan XY. Overexpression of HN1L promotes cell malignant proliferation in non-small cell lung cancer. Cancer Biol Ther 2017; 18:904-915. [PMID: 29053395 DOI: 10.1080/15384047.2017.1385678] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is a progressive malignant disease, involving the activation of oncogenes and inactivation of tumor suppressors. In this study, we identified and characterized a novel oncogene hematopoietic- and neurologic-expressed sequence 1-like (HN1L) in human NSCLC. Overexpression of HN1L was frequently detected in primary NSCLC compared with their non-tumor counterparts (P < 0.001), which was significantly associated with tumor size (P = 0.022). In addition, Kaplan-Meier analysis showed that upregulation of HN1L correlated with worse overall survival (P = 0.029) and disease-free survival (P = 0.011) for NSCLC patients. Both in vitro and in vivo studies demonstrated that inhibition of HN1L expression with shRNA dramatically inhibited cell growth, adherent and non-adherent colony formation, and tumorigenicity in nude mice. The positive correlation of HN1L expression and Ki67 level in a large NSCLC samples further suggested the key role of HN1L in the regulation of cell growth. Further study showed that knockdown of HN1L resulted in dramatic cell cycle arrest by interfering with MAPK pathway via interacting with RASA4 protein. In conclusion, HN1L plays a crucial role in the progression of NSCLC by contributing to malignant proliferation, with possible use as a new intervention point for therapeutic strategies.
Collapse
Affiliation(s)
- Lei Li
- a State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center , Guangzhou , Guangdong , China
| | - Ting-Ting Zeng
- a State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center , Guangzhou , Guangdong , China
| | - Bao-Zhu Zhang
- a State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center , Guangzhou , Guangdong , China
| | - Yan Li
- a State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center , Guangzhou , Guangdong , China
| | - Ying-Hui Zhu
- a State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center , Guangzhou , Guangdong , China
| | - Xin-Yuan Guan
- a State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center , Guangzhou , Guangdong , China.,b Department of Clinical Oncology , The University of Hong Kong , Hong Kong , China
| |
Collapse
|
13
|
Shah B, Püschel AW. Regulation of Rap GTPases in mammalian neurons. Biol Chem 2017; 397:1055-69. [PMID: 27186679 DOI: 10.1515/hsz-2016-0165] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 05/06/2016] [Indexed: 12/15/2022]
Abstract
Small GTPases are central regulators of many cellular processes. The highly conserved Rap GTPases perform essential functions in the mammalian nervous system during development and in mature neurons. During neocortical development, Rap1 is required to regulate cadherin- and integrin-mediated adhesion. In the adult nervous system Rap1 and Rap2 regulate the maturation and plasticity of dendritic spine and synapses. Although genetic studies have revealed important roles of Rap GTPases in neurons, their regulation by guanine nucleotide exchange factors (GEFs) that activate them and GTPase activating proteins (GAPs) that inactivate them by stimulating their intrinsic GTPase activity is just beginning to be explored in vivo. Here we review how GEFs and GAPs regulate Rap GTPases in the nervous system with a focus on their in vivo function.
Collapse
|
14
|
Wang H, Klein MG, Snell G, Lane W, Zou H, Levin I, Li K, Sang BC. Structure of Human GIVD Cytosolic Phospholipase A2 Reveals Insights into Substrate Recognition. J Mol Biol 2016; 428:2769-79. [PMID: 27220631 DOI: 10.1016/j.jmb.2016.05.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 05/09/2016] [Accepted: 05/13/2016] [Indexed: 11/18/2022]
Abstract
Cytosolic phospholipases A2 (cPLA2s) consist of a family of calcium-sensitive enzymes that function to generate lipid second messengers through hydrolysis of membrane-associated glycerophospholipids. The GIVD cPLA2 (cPLA2δ) is a potential drug target for developing a selective therapeutic agent for the treatment of psoriasis. Here, we present two X-ray structures of human cPLA2δ, capturing an apo state, and in complex with a substrate-like inhibitor. Comparison of the apo and inhibitor-bound structures reveals conformational changes in a flexible cap that allows the substrate to access the relatively buried active site, providing new insight into the mechanism for substrate recognition. The cPLA2δ structure reveals an unexpected second C2 domain that was previously unrecognized from sequence alignments, placing cPLA2δ into the class of membrane-associated proteins that contain a tandem pair of C2 domains. Furthermore, our structures elucidate novel inter-domain interactions and define three potential calcium-binding sites that are likely important for regulation and activation of enzymatic activity. These findings provide novel insights into the molecular mechanisms governing cPLA2's function in signal transduction.
Collapse
Affiliation(s)
- Hui Wang
- Department of Structural Biology, Takeda California, San Diego, CA 92121, USA.
| | - Michael G Klein
- Department of Structural Biology, Takeda California, San Diego, CA 92121, USA.
| | - Gyorgy Snell
- Department of Structural Biology, Takeda California, San Diego, CA 92121, USA
| | - Weston Lane
- Department of Structural Biology, Takeda California, San Diego, CA 92121, USA
| | - Hua Zou
- Department of Structural Biology, Takeda California, San Diego, CA 92121, USA
| | - Irena Levin
- Department of Structural Biology, Takeda California, San Diego, CA 92121, USA
| | - Ke Li
- Department of Structural Biology, Takeda California, San Diego, CA 92121, USA
| | - Bi-Ching Sang
- Department of Structural Biology, Takeda California, San Diego, CA 92121, USA
| |
Collapse
|
15
|
Strazza M, Azoulay-Alfaguter I, Dun B, Baquero-Buitrago J, Mor A. CD28 inhibits T cell adhesion by recruiting CAPRI to the plasma membrane. THE JOURNAL OF IMMUNOLOGY 2015; 194:2871-7. [PMID: 25637021 DOI: 10.4049/jimmunol.1401492] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CD28 is a coreceptor expressed on T lymphocytes. Signaling downstream of CD28 promotes multiple T cell functions such as proliferation, survival, and cytokine secretion. Adhesion to APCs is another function of T cells; however, little is known with regard to the role of CD28 in this process. Our previous studies have shown that CD28 inhibits T cell adhesion, but the underlying mechanism that mediates this process remains unknown. In the present study we discovered that signaling downstream of CD28 resulted in inhibition of Rap1 activity and decreased LFA-1-mediated adhesion. We showed that this was regulated by the recruitment of calcium-promoted Ras inactivator (CAPRI), a GTPase-activating protein, to the plasma membrane downstream of CD28 signaling. CAPRI trafficking to the plasma membrane was secondary to calcium influx and was mediated by its C2A and C2B domains. We conclude that CD28 inhibits Rap1-mediated adhesion by recruiting the protein CAPRI to the plasma membrane.
Collapse
Affiliation(s)
- Marianne Strazza
- Department of Medicine, New York University School of Medicine, New York, NY 10016
| | | | - Bryan Dun
- Department of Medicine, New York University School of Medicine, New York, NY 10016
| | | | - Adam Mor
- Department of Medicine, New York University School of Medicine, New York, NY 10016
| |
Collapse
|
16
|
Schurmans S, Polizzi S, Scoumanne A, Sayyed S, Molina-Ortiz P. The Ras/Rap GTPase activating protein RASA3: from gene structure to in vivo functions. Adv Biol Regul 2014; 57:153-61. [PMID: 25294679 DOI: 10.1016/j.jbior.2014.09.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 09/01/2014] [Accepted: 09/02/2014] [Indexed: 01/28/2023]
Abstract
RASA3 (or GTPase Activating Protein III, R-Ras GTPase-activating protein, GAP1(IP4BP)) is a GTPase activating protein of the GAP1 subfamily which targets Ras and Rap1. RASA3 was originally purified from pig platelet membranes through its intrinsic ability to bind inositol 1,3,4,5-tetrakisphosphate (I(1,3,4,5)P4) with high affinity, hence its first name GAP1(IP4BP) (for GAP1 subfamily member which binds I(1,3,4,5)P4). RASA3 was thus the first I(1,3,4,5)P4 receptor identified and cloned. The in vitro and in vivo functions of RASA3 remained somewhat elusive for a long time. However, recently, using genetically-modified mice and cells derived from these mice, the function of RASA3 during megakaryopoiesis, megakaryocyte adhesion and migration as well as integrin signaling has been reported. The goal of this review is thus to summarize and comment recent and less recent data in the literature on RASA3, in particular on the in vivo function of this specific GAP1 subfamily member.
Collapse
Affiliation(s)
- Stéphane Schurmans
- Laboratoire de Génétique Fonctionnelle, GIGA-Signal Transduction, GIGA B34, Université de Liège, Avenue de l'Hôpital 1, B-4000 Liège, Belgium; Secteur de Biochimie Métabolique, Département des Sciences Fonctionnelles, Faculté de Médecine Vétérinaire, Université de Liège, Boulevard de Colonster 20, 4000 Liège, Belgium; Welbio, Belgium.
| | - Séléna Polizzi
- Institut de Recherches Interdisciplinaires en Biologie Humaine et Moléculaire (IRIBHM), Institut de Biologie et de Médecine Moléculaires (IBMM), Faculté de Médecine, Université Libre de Bruxelles, Rue des Professeurs Jeener et Brachet 12, 6041 Gosselies, Belgium.
| | - Ariane Scoumanne
- Laboratoire de Génétique Fonctionnelle, GIGA-Signal Transduction, GIGA B34, Université de Liège, Avenue de l'Hôpital 1, B-4000 Liège, Belgium; Welbio, Belgium
| | - Sufyan Sayyed
- Laboratoire de Génétique Fonctionnelle, GIGA-Signal Transduction, GIGA B34, Université de Liège, Avenue de l'Hôpital 1, B-4000 Liège, Belgium
| | - Patricia Molina-Ortiz
- Laboratoire de Génétique Fonctionnelle, GIGA-Signal Transduction, GIGA B34, Université de Liège, Avenue de l'Hôpital 1, B-4000 Liège, Belgium; Welbio, Belgium
| |
Collapse
|
17
|
Poetsch AR, Lipka DB, Witte T, Claus R, Nöllke P, Zucknick M, Olk-Batz C, Fluhr S, Dworzak M, De Moerloose B, Starý J, Zecca M, Hasle H, Schmugge M, van den Heuvel-Eibrink MM, Locatelli F, Niemeyer CM, Flotho C, Plass C. RASA4 undergoes DNA hypermethylation in resistant juvenile myelomonocytic leukemia. Epigenetics 2014; 9:1252-60. [PMID: 25147919 DOI: 10.4161/epi.29941] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Aberrant DNA methylation at specific genetic loci is a key molecular feature of juvenile myelomonocytic leukemia (JMML) with poor prognosis. Using quantitative high-resolution mass spectrometry, we identified RASA4 isoform 2, which maps to chromosome 7 and encodes a member of the GAP1 family of GTPase-activating proteins for small G proteins, as a recurrent target of isoform-specific DNA hypermethylation in JMML (51% of 125 patients analyzed). RASA4 isoform 2 promoter methylation correlated with clinical parameters predicting poor prognosis (older age, elevated fetal hemoglobin), with higher risk of relapse after hematopoietic stem cell transplantation, and with PTPN11 mutation. The level of isoform 2 methylation increased in relapsed cases after transplantation. Interestingly, most JMML cases with monosomy 7 exhibited hypermethylation on the remaining RASA4 allele. The results corroborate the significance of epigenetic modifications in the phenotype of aggressive JMML.
Collapse
Affiliation(s)
- Anna R Poetsch
- Division of Epigenomics and Cancer Risk Factors (C010); German Cancer Research Center; Heidelberg, Germany
| | - Daniel B Lipka
- Division of Epigenomics and Cancer Risk Factors (C010); German Cancer Research Center; Heidelberg, Germany
| | - Tania Witte
- Division of Epigenomics and Cancer Risk Factors (C010); German Cancer Research Center; Heidelberg, Germany
| | - Rainer Claus
- Division of Epigenomics and Cancer Risk Factors (C010); German Cancer Research Center; Heidelberg, Germany
| | - Peter Nöllke
- Division of Pediatric Hematology-Oncology; University Medical Center; Freiburg, Germany
| | - Manuela Zucknick
- Division of Biostatistics; German Cancer Research Center; Heidelberg, Germany
| | - Christiane Olk-Batz
- Division of Pediatric Hematology-Oncology; University Medical Center; Freiburg, Germany
| | - Silvia Fluhr
- Division of Pediatric Hematology-Oncology; University Medical Center; Freiburg, Germany
| | - Michael Dworzak
- St. Anna Children's Hospital and Children's Cancer Research Institute; Department of Pediatrics; Medical University of Vienna; Vienna, Austria
| | - Barbara De Moerloose
- Dept. of Pediatric Hematology-Oncology; Ghent University Hospital; Ghent, Belgium
| | - Jan Starý
- Dept. of Pediatric Hematology and Oncology; 2nd Faculty of Medicine; Charles University and University Hospital Motol, Praha; Czech Pediatric Hematology Working Group (CPH); Prague, Czech Republic
| | - Marco Zecca
- Pediatric Hematology-Oncology; Fondazione IRCCS;Policlinico San Matteo; Pavia, Italy
| | - Henrik Hasle
- Department of Pediatrics; Aarhus University Hospital Skejby; Aarhus, Denmark
| | - Markus Schmugge
- Division of Hematology; University Children's Hospital; Zurich, Switzerland
| | - Marry M van den Heuvel-Eibrink
- Department of Pediatric Oncology-Hematology; Erasmus Medical Center; Rotterdam, The Netherlands; Dutch Children's Oncology Group; The Hague, Netherlands
| | - Franco Locatelli
- Department of Pediatric Hematology-Oncology; IRCCS Ospedale Bambino Gesu; Rome, Italy; University of Pavia; Pavia, Italy
| | - Charlotte M Niemeyer
- The German Cancer Consortium; Heidelberg, Germany; Division of Pediatric Hematology-Oncology; University Medical Center; Freiburg, Germany
| | - Christian Flotho
- The German Cancer Consortium; Heidelberg, Germany; Division of Pediatric Hematology-Oncology; University Medical Center; Freiburg, Germany
| | - Christoph Plass
- Division of Epigenomics and Cancer Risk Factors (C010); German Cancer Research Center; Heidelberg, Germany; The German Cancer Consortium; Heidelberg, Germany
| |
Collapse
|
18
|
Molina-Ortiz P, Polizzi S, Ramery E, Gayral S, Delierneux C, Oury C, Iwashita S, Schurmans S. Rasa3 controls megakaryocyte Rap1 activation, integrin signaling and differentiation into proplatelet. PLoS Genet 2014; 10:e1004420. [PMID: 24967784 PMCID: PMC4072513 DOI: 10.1371/journal.pgen.1004420] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Accepted: 04/20/2014] [Indexed: 01/17/2023] Open
Abstract
Rasa3 is a GTPase activating protein of the GAP1 family which targets Ras and Rap1. Ubiquitous Rasa3 catalytic inactivation in mouse results in early embryonic lethality. Here, we show that Rasa3 catalytic inactivation in mouse hematopoietic cells results in a lethal syndrome characterized by severe defects during megakaryopoiesis, thrombocytopenia and a predisposition to develop preleukemia. The main objective of this study was to define the cellular and the molecular mechanisms of terminal megakaryopoiesis alterations. We found that Rasa3 catalytic inactivation altered megakaryocyte development, adherence, migration, actin cytoskeleton organization and differentiation into proplatelet forming megakaryocytes. These megakaryocyte alterations were associated with an increased active Rap1 level and a constitutive integrin activation. Thus, these mice presented a severe thrombocytopenia, bleeding and anemia associated with an increased percentage of megakaryocytes in the bone marrow, bone marrow fibrosis, extramedular hematopoiesis, splenomegaly and premature death. Altogether, our results indicate that Rasa3 catalytic activity controls Rap1 activation and integrin signaling during megakaryocyte differentiation in mouse. Megakaryocytes are the bone marrow cellular precursors of circulating blood platelets and give rise to nascent platelets by forming branching filaments called proplatelets. Terminal differentiation of round megakaryocytes into branched proplatelet forming megakaryocytes is a complex cytoskeletal-driven process which is affected in rare human familial thrombocytopenias. Interactions of megakaryocytes with extracellular matrix proteins are essential in this process since constitutive megakaryocyte integrin activity caused by specific mutations in ITGA2B or ITGB3 genes encoding for extracellular matrix protein receptors may result in abnormal adherent megakaryocytes, defect in proplatelet formation and thrombocytopenia. Here, we show that Rasa3, a GTPase activating protein of the GAP1 family, controls Rap1 activation and integrin signaling during megakaryocyte differentiation. We found that Rasa3 catalytic inactivation in mice altered megakaryocyte development, adherence, migration, actin cytoskeleton organization and differentiation into proplatelet. Thus, these mice presented a severe thrombocytopenia, bleeding and anemia.
Collapse
Affiliation(s)
- Patricia Molina-Ortiz
- Laboratory of Functional Genetics, GIGA-Research Centre, Université de Liège, Liège, and Welbio, Belgium
| | - Séléna Polizzi
- Institut de Recherches Interdisciplinaires en Biologie Humaine et Moléculaire (IRIBHM), Institut de Biologie et de Médecine Moléculaires (IBMM), Faculté de Médecine, Université Libre de Bruxelles, Gosselies, Belgium
| | - Eve Ramery
- Laboratoire de Biologie Clinique, Faculté de Médecine-vétérinaire, Université de Liège, Liège, Belgium
| | - Stéphanie Gayral
- Institut de Recherches Interdisciplinaires en Biologie Humaine et Moléculaire (IRIBHM), Institut de Biologie et de Médecine Moléculaires (IBMM), Faculté de Médecine, Université Libre de Bruxelles, Gosselies, Belgium
| | - Céline Delierneux
- Laboratory of Thrombosis and Hemostasis, GIGA-Research Centre, Université de Liège, Liège, Belgium
| | - Cécile Oury
- Laboratory of Thrombosis and Hemostasis, GIGA-Research Centre, Université de Liège, Liège, Belgium
| | - Shintaro Iwashita
- Mitsubishi Kagaku Institute of Life Sciences and Faculty of Pharmacy, Iwaki Meisei University, Iwaki, Japan
| | - Stéphane Schurmans
- Laboratory of Functional Genetics, GIGA-Research Centre, Université de Liège, Liège, and Welbio, Belgium
- Institut de Recherches Interdisciplinaires en Biologie Humaine et Moléculaire (IRIBHM), Institut de Biologie et de Médecine Moléculaires (IBMM), Faculté de Médecine, Université Libre de Bruxelles, Gosselies, Belgium
- * E-mail:
| |
Collapse
|
19
|
Frequency decoding of calcium oscillations. Biochim Biophys Acta Gen Subj 2014; 1840:964-9. [DOI: 10.1016/j.bbagen.2013.11.015] [Citation(s) in RCA: 179] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 09/27/2013] [Accepted: 11/15/2013] [Indexed: 01/14/2023]
|
20
|
Akiyama H, Kamiguchi H. Second messenger networks for accurate growth cone guidance. Dev Neurobiol 2013; 75:411-22. [PMID: 24285606 DOI: 10.1002/dneu.22157] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 11/21/2013] [Accepted: 11/25/2013] [Indexed: 02/02/2023]
Abstract
Growth cones are able to navigate over long distances to find their appropriate target by following guidance cues that are often presented to them in the form of an extracellular gradient. These external cues are converted into gradients of specific signaling molecules inside growth cones, while at the same time these internal signals are amplified. The amplified instruction is then used to generate asymmetric changes in the growth cone turning machinery so that one side of the growth cone migrates at a rate faster than the other side, and thus the growth cone turns toward or away from the external cue. This review examines how signal specification and amplification can be achieved inside the growth cone by multiple second messenger signaling pathways activated downstream of guidance cues. These include the calcium ion, cyclic nucleotide, and phosphatidylinositol signaling pathways.
Collapse
Affiliation(s)
- Hiroki Akiyama
- Laboratory for Neuronal Growth Mechanisms, RIKEN Brain Science Institute, Wako, Saitama, 351-0198, Japan
| | | |
Collapse
|
21
|
Brownlie RJ, Zamoyska R. T cell receptor signalling networks: branched, diversified and bounded. Nat Rev Immunol 2013; 13:257-69. [PMID: 23524462 DOI: 10.1038/nri3403] [Citation(s) in RCA: 339] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Engagement of antigen-specific T cell receptors (TCRs) is a prerequisite for T cell activation. Acquisition of appropriate effector T cell function requires the participation of multiple signals from the T cell microenvironment. Trying to understand how these signals integrate to achieve specific functional outcomes while maintaining tolerance to self is a major challenge in lymphocyte biology. Several recent publications have provided important insights into how dysregulation of T cell signalling and the development of autoreactivity can result if the branching and integration of signalling pathways are perturbed. We discuss how these findings highlight the importance of spatial segregation of individual signalling components as a way of regulating T cell responsiveness and immune tolerance.
Collapse
Affiliation(s)
- Rebecca J Brownlie
- Institute for Immunology and Infection Research, The University of Edinburgh, Edinburgh EH9 3JT, UK
| | | |
Collapse
|
22
|
Idevall-Hagren O, Jakobsson I, Xu Y, Tengholm A. Spatial control of Epac2 activity by cAMP and Ca2+-mediated activation of Ras in pancreatic β cells. Sci Signal 2013; 6:ra29.1-11, S1-6. [PMID: 23633676 DOI: 10.1126/scisignal.2003932] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The cAMP (adenosine 3',5'-monophosphate)-activated guanine nucleotide exchange factor (GEF) Epac2 is an important mediator of cAMP-dependent processes in multiple cell types. We used real-time confocal and total internal reflection fluorescence microscopy to examine the spatiotemporal regulation of Epac2, which is a GEF for the guanosine triphosphatase (GTPase) Rap. We demonstrated that increases in the concentration of cAMP triggered the translocation of Epac2 from the cytoplasm to the plasma membrane in insulin-secreting β cells. Glucose-induced oscillations of the submembrane concentration of cAMP were associated with cyclic translocation of Epac2, and this translocation could be amplified by increases in the cytoplasmic Ca(2+) concentration. Analyses of Epac2 mutants identified the high-affinity cAMP-binding and the Ras association domains as crucial for the translocation. Expression of a dominant-negative Ras mutant reduced Epac2 translocation, and Ca(2+)-dependent oscillations in Ras activity synchronized with Epac2 translocation in single β cells. The cyclic translocation of Epac2 was accompanied by oscillations of Rap GTPase activity at the plasma membrane, and expression of an inactive Rap1B mutant decreased insulin secretion. Thus, Epac2 localization is dynamically controlled by cAMP as well as by Ca(2+)-mediated activation of Ras. These results help to explain how oscillating signals can produce pulses of insulin release from pancreatic β cells.
Collapse
Affiliation(s)
- Olof Idevall-Hagren
- Department of Medical Cell Biology, Uppsala University, Biomedical Centre, Box 571, SE-751 23 Uppsala, Sweden
| | | | | | | |
Collapse
|
23
|
King PD, Lubeck BA, Lapinski PE. Nonredundant functions for Ras GTPase-activating proteins in tissue homeostasis. Sci Signal 2013; 6:re1. [PMID: 23443682 DOI: 10.1126/scisignal.2003669] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Inactivation of the small guanosine triphosphate-binding protein Ras during receptor signal transduction is mediated by Ras guanosine triphosphatase (GTPase)-activating proteins (RasGAPs). Ten different RasGAPs have been identified and have overlapping patterns of tissue distribution. However, genetic analyses are revealing critical nonredundant functions for each RasGAP in tissue homeostasis and as regulators of disease processes in mouse and man. Here, we discuss advances in understanding the role of RasGAPs in the maintenance of tissue integrity.
Collapse
Affiliation(s)
- Philip D King
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | | | | |
Collapse
|
24
|
Ras GTPase activating (RasGAP) activity of the dual specificity GAP protein Rasal requires colocalization and C2 domain binding to lipid membranes. Proc Natl Acad Sci U S A 2012; 110:111-6. [PMID: 23251034 DOI: 10.1073/pnas.1201658110] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Rasal, belonging to the GAP1 subfamily of Ras GTPase-activating proteins (RasGAPs) with dual RasGAP/RapGAP specificity, is epigenetically silenced in several tumor types. Surprisingly, the isolated protein has GAP activity on Rap but not on Ras. Its membrane recruitment is regulated by interaction with calcium and lipids, which simultaneously induces its RasGAP activity through a yet unknown mechanism. Here we show that the interaction of Rasal with membranes induces Rasal RasGAP activity by spatial and conformational regulation, although it does not have any effect on its RapGAP activity. Not only is colocalization of Rasal and Ras in the membrane essential for RasGAP activation, but direct and Ca-dependent interaction between the tandem C2 domains of Rasal and lipids of the membrane is also required. Whereas the C2A domain binds specifically phosphatidylserine, the C2B domain interacts with several phosphoinositol lipids. Finally we show, that similar to the C2 domains of synaptotagmins, the Rasal tandem C2 domains are able to sense and induce membrane curvature by the insertion of hydrophobic loops into the membrane.
Collapse
|
25
|
Abstract
Ever since their discovery as cellular counterparts of viral oncogenes more than 25 years ago, much progress has been made in understanding the complex networks of signal transduction pathways activated by oncogenic Ras mutations in human cancers. The activity of Ras is regulated by nucleotide exchange factors (GEFs) and GTPase activating proteins (GAPs), and much emphasis has been put into the biochemical and structural analysis of the Ras/GAP complex. The mechanisms by which GAPs catalyze Ras-GTP hydrolysis have been clarified and revealed that oncogenic Ras mutations confer resistance to GAPs and remain constitutively active. However, it is yet unclear how cells coordinate the large and divergent GAP protein family to promote Ras inactivation and ensure a certain biological response. Different domain arrangements in GAPs to create differential protein-protein and protein-lipid interactions are probably key factors determining the inactivation of the 3 Ras isoforms H-, K-, and N-Ras and their effector pathways. In recent years, in vitro as well as cell- and animal-based studies examining GAP activity, localization, interaction partners, and expression profiles have provided further insights into Ras inactivation and revealed characteristics of several GAPs to exert specific and distinct functions. This review aims to summarize knowledge on the cell biology of RasGAP proteins that potentially contributes to differential regulation of spatiotemporal Ras signaling.
Collapse
Affiliation(s)
- Thomas Grewal
- Faculty of Pharmacy, University of Sydney, Sydney, NSW, Australia
| | | | | | | |
Collapse
|
26
|
Steininger A, Möbs M, Ullmann R, Köchert K, Kreher S, Lamprecht B, Anagnostopoulos I, Hummel M, Richter J, Beyer M, Janz M, Klemke CD, Stein H, Dörken B, Sterry W, Schrock E, Mathas S, Assaf C. Genomic loss of the putative tumor suppressor gene E2A in human lymphoma. ACTA ACUST UNITED AC 2011; 208:1585-93. [PMID: 21788410 PMCID: PMC3149217 DOI: 10.1084/jem.20101785] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
The transcription factor E2A is essential for lymphocyte development. In this study, we describe a recurrent E2A gene deletion in at least 70% of patients with Sézary syndrome (SS), a subtype of T cell lymphoma. Loss of E2A results in enhanced proliferation and cell cycle progression via derepression of the protooncogene MYC and the cell cycle regulator CDK6. Furthermore, by examining the gene expression profile of SS cells after restoration of E2A expression, we identify several E2A-regulated genes that interfere with oncogenic signaling pathways, including the Ras pathway. Several of these genes are down-regulated or lost in primary SS tumor cells. These data demonstrate a tumor suppressor function of E2A in human lymphoid cells and could help to develop new treatment strategies for human lymphomas with altered E2A activity.
Collapse
Affiliation(s)
- Anne Steininger
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Rovillain E, Mansfield L, Lord CJ, Ashworth A, Jat PS. An RNA interference screen for identifying downstream effectors of the p53 and pRB tumour suppressor pathways involved in senescence. BMC Genomics 2011; 12:355. [PMID: 21740549 PMCID: PMC3161017 DOI: 10.1186/1471-2164-12-355] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Accepted: 07/08/2011] [Indexed: 12/28/2022] Open
Abstract
Background Cellular senescence is an irreversible cell cycle arrest that normal cells undergo in response to progressive shortening of telomeres, changes in telomeric structure, oncogene activation or oxidative stress and acts as an important tumour suppressor mechanism. Results To identify the downstream effectors of the p53-p21 and p16-pRB tumour suppressor pathways crucial for mediating entry into senescence, we have carried out a loss-of-function RNA interference screen in conditionally immortalised human fibroblasts that can be induced to rapidly undergo senescence, whereas in primary cultures senescence is stochastic and occurs asynchronously. These cells are immortal but undergo a rapid irreversible arrest upon activation of the p53-p21 and p16-pRB pathways that can be readily bypassed upon their inactivation. The primary screen identified 112 known genes including p53 and another 29 shRNAmirs targetting as yet unidentified loci. Comparison of these known targets with genes known to be up-regulated upon senescence in these cells, by micro-array expression profiling, identified 4 common genes TMEM9B, ATXN10, LAYN and LTBP2/3. Direct silencing of these common genes, using lentiviral shRNAmirs, bypassed senescence in the conditionally immortalised cells. Conclusion The senescence bypass screen identified TMEM9B, ATXN10, LAYN and LTBP2/3 as novel downstream effectors of the p53-p21 and p16-pRB tumour suppressor pathways. Although none of them has previously been linked to cellular senescence, TMEM9B has been suggested to be an upstream activator of NF-κB signalling which has been found to have a causal role in promoting senescence. Future studies will focus on determining on how many of the other primary hits also have a casual role in senescence and what is the mechanism of action.
Collapse
Affiliation(s)
- Emilie Rovillain
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | | | | | | | | |
Collapse
|
28
|
Dai Y, Walker SA, de Vet E, Cook S, Welch HCE, Lockyer PJ. Ca2+-dependent monomer and dimer formation switches CAPRI Protein between Ras GTPase-activating protein (GAP) and RapGAP activities. J Biol Chem 2011; 286:19905-16. [PMID: 21460216 DOI: 10.1074/jbc.m110.201301] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
CAPRI is a member of the GAP1 family of GTPase-activating proteins (GAPs) for small G proteins. It is known to function as an amplitude sensor for intracellular Ca(2+) levels stimulated by extracellular signals and has a catalytic domain with dual RasGAP and RapGAP activities. Here, we have investigated the mechanism that switches CAPRI between its two GAP activities. We demonstrate that CAPRI forms homodimers in vitro and in vivo in a Ca(2+)-dependent manner. The site required for dimerization was pinpointed by deletion and point mutations to a helix motif that forms a hydrophobic face in the extreme C-terminal tail of the CAPRI protein. Deletion of this helix motif abolished dimer formation but did not affect translocation of CAPRI to the plasma membrane upon cell stimulation with histamine. We found that dimeric and monomeric CAPRI coexist in cells and that the ratio of dimeric to monomeric CAPRI increases upon cell stimulation with histamine. Free Ca(2+) at physiologically relevant concentrations was both necessary and sufficient for dimer formation. Importantly, the monomeric and dimeric forms of CAPRI exhibited differential GAP activities in vivo; the wild-type form of CAPRI had stronger RapGAP activity than RasGAP activity, whereas a monomeric CAPRI mutant showed stronger RasGAP than RapGAP activity. These results demonstrate that CAPRI switches between its dual GAP roles by forming monomers or homodimers through a process regulated by Ca(2+). We propose that Ca(2+)-dependent dimerization of CAPRI may serve to coordinate Ras and Rap1 signaling pathways.
Collapse
Affiliation(s)
- Yanfeng Dai
- Laboratory of Molecular Signalling, The Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom.
| | | | | | | | | | | |
Collapse
|
29
|
Lorentzen A, Kinkhabwala A, Rocks O, Vartak N, Bastiaens PIH. Regulation of Ras localization by acylation enables a mode of intracellular signal propagation. Sci Signal 2010; 3:ra68. [PMID: 20858867 DOI: 10.1126/scisignal.20001370] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Growth factor stimulation generates transient H-Ras activity at the plasma membrane but sustained activity at the Golgi. Two overlapping regulatory networks control compartmentalized H-Ras activity: the guanosine diphosphate-guanosine triphosphate cycle and the acylation cycle, which constitutively traffics Ras isoforms that can be palmitoylated between intracellular membrane compartments. Quantitative imaging of H-Ras activity after decoupling of these networks revealed regulation of H-Ras activity at the plasma membrane but not at the Golgi. Nevertheless, upon stimulation with epidermal growth factor, Ras activity at the Golgi displayed a pulse-like profile similar to that at the plasma membrane but also remained high after the initial stimulus. A compartmental model that included the acylation cycle and H-Ras regulation at the plasma membrane accounted for the pulse-like profile of H-Ras activity at the Golgi but implied that sustained H-Ras activity at the Golgi required H-Ras activation at an additional compartment, which we experimentally determined to be the endoplasmic reticulum. Thus, in addition to maintaining the localization of Ras, the acylation cycle underlies a previously unknown form of signal propagation similar to radio transmission in its generation of a constitutive Ras "carrier wave" that transmits Ras activity between subcellular compartments.
Collapse
Affiliation(s)
- Anna Lorentzen
- European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | | | | | | | | |
Collapse
|
30
|
Colombo S, Kumasaka M, Lobe C, Larue L. Genomic localization of the Z/EG transgene in the mouse genome. Genesis 2010; 48:96-100. [PMID: 20014334 DOI: 10.1002/dvg.20585] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The Z/EG transgenic mouse line, produced by Novak et al., displays tissue-specific EGFP expression after Cre-mediated recombination. The autofluorescence of EGFP allows the visualization of cells of interest displaying Cre recombination. The initial construct was designed such that cells without Cre recombination express the beta-galactosidase marker, facilitating counterselection. We used inverse PCR to identify the site of integration of the Z/EG transgene, to improve the efficiency of homozygous Z/EG mouse production. Recombined cells produced large amounts of EGFP protein, resulting in higher levels of fluorescence and therefore greater contrast with nonrecombined cells. We mapped the transgene to the G1 region of chromosome 5. This random insertion was found to have occurred 230-bp upstream from the start codon of the Rasa4 gene. The insertion of the Z/EG transgene in the C57BL/6 genetic background had no effect on Rasa4 expression. Homozygous Z/EG mice therefore had no obvious phenotype.
Collapse
Affiliation(s)
- Sophie Colombo
- Institut Curie, Centre de Recherche, Developmental Genetics of Melanocytes, 91405, Orsay, France
| | | | | | | |
Collapse
|
31
|
Allosteric modulation of Ras positions Q61 for a direct role in catalysis. Proc Natl Acad Sci U S A 2010; 107:4931-6. [PMID: 20194776 DOI: 10.1073/pnas.0912226107] [Citation(s) in RCA: 197] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Ras and its effector Raf are key mediators of the Ras/Raf/MEK/ERK signal transduction pathway. Mutants of residue Q61 impair the GTPase activity of Ras and are found prominently in human cancers. Yet the mechanism through which Q61 contributes to catalysis has been elusive. It is thought to position the catalytic water molecule for nucleophilic attack on the gamma-phosphate of GTP. However, we previously solved the structure of Ras from crystals with symmetry of the space group R32 in which switch II is disordered and found that the catalytic water molecule is present. Here we present a structure of wild-type Ras with calcium acetate from the crystallization mother liquor bound at a site remote from the active site and likely near the membrane. This results in a shift in helix 3/loop 7 and a network of H-bonding interactions that propagates across the molecule, culminating in the ordering of switch II and placement of Q61 in the active site in a previously unobserved conformation. This structure suggests a direct catalytic role for Q61 where it interacts with a water molecule that bridges one of the gamma-phosphate oxygen atoms to the hydroxyl group of Y32 to stabilize the transition state of the hydrolysis reaction. We propose that Raf together with the binding of Ca(2+) and a negatively charged group mimicked in our structure by the acetate molecule induces the ordering of switch I and switch II to complete the active site of Ras.
Collapse
|
32
|
Priatel JJ, Chen X, Huang YH, Chow MT, Zenewicz LA, Coughlin JJ, Shen H, Stone JC, Tan R, Teh HS. RasGRP1 regulates antigen-induced developmental programming by naive CD8 T cells. THE JOURNAL OF IMMUNOLOGY 2009; 184:666-76. [PMID: 20007535 DOI: 10.4049/jimmunol.0803521] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Ag encounter by naive CD8 T cells initiates a developmental program consisting of cellular proliferation, changes in gene expression, and the formation of effector and memory T cells. The strength and duration of TCR signaling are known to be important parameters regulating the differentiation of naive CD8 T cells, although the molecular signals arbitrating these processes remain poorly defined. The Ras-guanyl nucleotide exchange factor RasGRP1 has been shown to transduce TCR-mediated signals critically required for the maturation of developing thymocytes. To elucidate the role of RasGRP1 in CD8 T cell differentiation, in vitro and in vivo experiments were performed with 2C TCR transgenic CD8 T cells lacking RasGRP1. In this study, we report that RasGRP1 regulates the threshold of T cell activation and Ag-induced expansion, at least in part, through the regulation of IL-2 production. Moreover, RasGRP1(-/-) 2C CD8 T cells exhibit an anergic phenotype in response to cognate Ag stimulation that is partially reversible upon the addition of exogenous IL-2. By contrast, the capacity of IL-2/IL-2R interactions to mediate Ras activation and CD8 T cell expansion and differentiation appears to be largely RasGRP1-independent. Collectively, our results demonstrate that RasGRP1 plays a selective role in T cell signaling, controlling the initiation and duration of CD8 T cell immune responses.
Collapse
Affiliation(s)
- John J Priatel
- Child and Family Research Institute, Vancouver, British Columbia, Canada.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
The ability of GAP1IP4BP to function as a Rap1 GTPase-activating protein (GAP) requires its Ras GAP-related domain and an arginine finger rather than an asparagine thumb. Mol Cell Biol 2009; 29:3929-40. [PMID: 19433443 DOI: 10.1128/mcb.00427-09] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
GAP1(IP4BP) is a member of the GAP1 family of Ras GTPase-activating proteins (GAPs) that includes GAP1(m), CAPRI, and RASAL. Composed of a central Ras GAP-related domain (RasGRD), surrounded by amino-terminal C2 domains and a carboxy-terminal PH/Btk domain, these proteins, with the notable exception of GAP1(m), possess an unexpected arginine finger-dependent GAP activity on the Ras-related protein Rap1 (S. Kupzig, D. Deaconescu, D. Bouyoucef, S. A. Walker, Q. Liu, C. L. Polte, O. Daumke, T. Ishizaki, P. J. Lockyer, A. Wittinghofer, and P. J. Cullen, J. Biol. Chem. 281:9891-9900, 2006). Here, we have examined the mechanism through which GAP1(IP4BP) can function as a Rap1 GAP. We show that deletion of domains on either side of the RasGRD, while not affecting Ras GAP activity, do dramatically perturb Rap1 GAP activity. By utilizing GAP1(IP4BP)/GAP1(m) chimeras, we establish that although the C2 and PH/Btk domains are required to stabilize the RasGRD, it is this domain which contains the catalytic machinery required for Rap1 GAP activity. Finally, a key residue in Rap1-specific GAPs is a catalytic asparagine, the so-called asparagine thumb. By generating a molecular model describing the predicted Rap1-binding site in the RasGRD of GAP1(IP4BP), we show that mutagenesis of individual asparagine or glutamine residues that lie in close proximity to the predicted binding site has no detectable effect on the in vivo Rap1 GAP activity of GAP1(IP4BP). In contrast, we present evidence consistent with a model in which the RasGRD of GAP1(IP4BP) functions to stabilize the switch II region of Rap1, allowing stabilization of the transition state during GTP hydrolysis initiated by the arginine finger.
Collapse
|
34
|
Seidl C, Port M, Apostolidis C, Bruchertseifer F, Schwaiger M, Senekowitsch-Schmidtke R, Abend M. Differential gene expression triggered by highly cytotoxic alpha-emitter-immunoconjugates in gastric cancer cells. Invest New Drugs 2009; 28:49-60. [PMID: 19139817 DOI: 10.1007/s10637-008-9214-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2008] [Accepted: 12/19/2008] [Indexed: 12/20/2022]
Abstract
Immunoconjugates composed of the alpha-emitter (213)Bi and the monoclonal antibody d9MAb specifically target HSC45-M2 gastric cancer cells expressing mutant d9-E-cadherin. These conjugates efficiently killed tumor cells in a nude mouse peritoneal carcinomatosis model. To elucidate the molecular responses of HSC45-M2 cells to alpha-emitter irradiation, whole genome gene expression profiling was performed. For that purpose HSC45-M2 cells were incubated with lethal doses of (213)Bi-d9MAb. RNA was isolated at 6, 24 and 48 h after irradiation, transcribed into cDNA and hybridized to whole genome microarrays. Results of microarray analysis were validated using RTQ-PCR showing correspondence of approximately 90%. Following incubation with (213)Bi-d9MAb, 682-1125 genes showed upregulation and 666-1278 genes showed downregulation at one time point, each. Eight genes appeared upregulated and 12 genes downregulated throughout. Molecular functions and biological processes of differentially expressed genes were categorized according to the PANTHER database. Following (213)Bi-d9MAb irradiation also a time-dependent shift in terms of overrepresentation of biological processes was observed. Among the genes showing continuous upregulation, COL4A2, NEDD9 and C3 have not been associated with the cellular response to high LET radiation so far. The same holds true for WWP2, RFX3, HIST4H4 and JADE1 that showed continuous downregulation. According to PANTHER, three of the consistently upregulated (ITM2C, FLJ11000, MSMB) and downregulated (HCG9, GAS2L3, FLJ21439) genes, respectively, have not been associated with any biological process or molecular function so far. Thus, these findings revealed interesting new targets for selective elimination of tumor cells and new insights regarding response of tumor cells to alpha-emitter exposure.
Collapse
Affiliation(s)
- Christof Seidl
- Department of Nuclear Medicine, Technische Universität München, Ismaninger Strasse 22, 81675, Munich, Germany.
| | | | | | | | | | | | | |
Collapse
|
35
|
Hitomi J, Christofferson DE, Ng A, Yao J, Degterev A, Xavier RJ, Yuan J. Identification of a molecular signaling network that regulates a cellular necrotic cell death pathway. Cell 2009; 135:1311-23. [PMID: 19109899 DOI: 10.1016/j.cell.2008.10.044] [Citation(s) in RCA: 877] [Impact Index Per Article: 54.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2008] [Revised: 09/08/2008] [Accepted: 10/10/2008] [Indexed: 11/15/2022]
Abstract
Stimulation of death receptors by agonists such as FasL and TNFalpha activates apoptotic cell death in apoptotic-competent conditions or a type of necrotic cell death dependent on RIP1 kinase, termed necroptosis, in apoptotic-deficient conditions. In a genome-wide siRNA screen for regulators of necroptosis, we identify a set of 432 genes that regulate necroptosis, a subset of 32 genes that act downstream and/or as regulators of RIP1 kinase, 32 genes required for death-receptor-mediated apoptosis, and 7 genes involved in both necroptosis and apoptosis. We show that the expression of subsets of the 432 genes is enriched in the immune and nervous systems, and cellular sensitivity to necroptosis is regulated by an extensive signaling network mediating innate immunity. Interestingly, Bmf, a BH3-only Bcl-2 family member, is required for death-receptor-induced necroptosis. Our study defines a cellular signaling network that regulates necroptosis and the molecular bifurcation that controls apoptosis and necroptosis.
Collapse
Affiliation(s)
- Junichi Hitomi
- Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Origin of the sharp boundary that discriminates positive and negative selection of thymocytes. Proc Natl Acad Sci U S A 2008; 106:528-33. [PMID: 19098101 DOI: 10.1073/pnas.0805981105] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
T lymphocytes play a key role in adaptive immunity and are activated by interactions of their T cell receptors (TCR) with peptides (p) derived from antigenic proteins bound to MHC gene products. The repertoire of T lymphocytes available in peripheral organs is tuned in the thymus. Immature T lymphocytes (thymocytes) interact with diverse endogenous peptides bound to MHC in the thymus. TCR expressed on thymocytes must bind weakly to endogenous pMHC (positive selection) but must not bind too strongly to them (negative selection) to emerge from the thymus. Negatively selecting pMHC ligands bind TCR with a binding affinity that exceeds a sharply defined (digital) threshold. In contrast, there is no sharp threshold separating positively selecting ligands from those that bind too weakly to elicit a response. We describe results of computer simulations and experiments, which suggest that the contrast between the characters of the positive and negative selection thresholds originates in differences in the way in which Ras proteins are activated by ligands of varying potency. The molecular mechanism suggested by our studies generates hypotheses for how genetic aberrations may dampen the digital negative selection response, with concomitant escape of autoimmune T lymphocytes from the thymus.
Collapse
|
37
|
Pamonsinlapatham P, Hadj-Slimane R, Lepelletier Y, Allain B, Toccafondi M, Garbay C, Raynaud F. p120-Ras GTPase activating protein (RasGAP): a multi-interacting protein in downstream signaling. Biochimie 2008; 91:320-8. [PMID: 19022332 DOI: 10.1016/j.biochi.2008.10.010] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2008] [Accepted: 10/16/2008] [Indexed: 11/29/2022]
Abstract
p120-RasGAP (Ras GTPase activating protein) plays a key role in the regulation of Ras-GTP bound by promoting GTP hydrolysis via its C-terminal catalytic domain. The p120-RasGAP N-terminal part contains two SH2, SH3, PH (pleckstrin homology) and CaLB/C2 (calcium-dependent phospholipid-binding domain) domains. These protein domains allow various functions, such as anti-/pro-apoptosis, proliferation and also cell migration depending of their distinct partners. The p120-RasGAP domain participates in protein-protein interactions with Akt, Aurora or RhoGAP to regulate functions described bellow. Here, we summarize, in angiogenesis and cancer, the various functional roles played by p120-RasGAP domains and their effector partners in downstream signaling.
Collapse
Affiliation(s)
- Perayot Pamonsinlapatham
- Université Paris Descartes, UFR Biomédicale, Laboratoire de Pharmacochimie Moléculaire et Cellulaire, 45 Rue des Saints-Pères, 75270 Paris Cedex 06, France
| | | | | | | | | | | | | |
Collapse
|
38
|
Shemarova IV, Nesterov VP. Evolution of mechanisms of Ca2+-signaling. Role of Ca2+ in regulation of fundamental cell functions. J EVOL BIOCHEM PHYS+ 2008. [DOI: 10.1134/s0022093008040017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
39
|
Vilá de Muga S, Timpson P, Cubells L, Evans R, Hayes TE, Rentero C, Hegemann A, Reverter M, Leschner J, Pol A, Tebar F, Daly RJ, Enrich C, Grewal T. Annexin A6 inhibits Ras signalling in breast cancer cells. Oncogene 2008; 28:363-77. [PMID: 18850003 DOI: 10.1038/onc.2008.386] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Overexpression of epidermal growth factor receptor (EGFR) is associated with enhanced activation of wild-type (hyperactive) Ras in breast cancer. Little is known about the regulation of Ras inactivation and GTPase-activating proteins (GAPs), such as p120GAP, in cells with hyperactive Ras. Recently, we showed that in EGFR-overexpressing A431 cells, which lack endogenous Annexin A6 (AnxA6), ectopic expression of AnxA6 stimulates membrane recruitment of p120GAP to modulate Ras signalling. We now demonstrate that, AnxA6 is downregulated in a number of EGFR-overexpressing and estrogen receptor (ER)-negative breast cancer cells. In these cells, AnxA6 overexpression promotes Ca(2+)- and EGF-inducible membrane targeting of p120GAP. In ER-negative MDA-MB-436 cells, overexpression of p120GAP, but not CAPRI or a p120GAP mutant lacking the AnxA6-binding domain inhibits Ras/MAPK activity. AnxA6 knockdown in MDA-MB-436 increases Ras activity and cell proliferation in anchorage-independent growth assays. Furthermore, AnxA6 co-immunoprecipitates with H-Ras in a Ca(2+)- and EGF-inducible manner and fluorescence resonance energy transfer (FRET) microscopy confirmed that AnxA6 is in close proximity of active (G12V), but not inactive (S17N) H-Ras. Thus, association of AnxA6 with H-Ras-containing protein complexes may contribute to regulate p120GAP/Ras assembly in EGFR-overexpressing and ER-negative breast cancer cells.
Collapse
Affiliation(s)
- S Vilá de Muga
- Departament de Biologia Cellular, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
The mitogen-activated protein kinase (MAPK) pathway provides cells with the means to interpret external signal cues or conditions, and respond accordingly. This cascade regulates many cell functions such as differentiation, proliferation and migration. Through modulation of both the amplitude and duration of MAPK signalling, cells can control their responses to the multiple activators of the pathway. In addition, recent work has highlighted the importance of the cellular compartment from which the signalling occurs. Cells have developed intricate systems that enable them to localise MAPK components to specific subcellular domains in response to a particular stimulus. Consequently, different factors can activate the same kinase in separate locations. Crucial to this ability are molecular scaffolds, which act as signalling modules for MAPKs, confining them to the desired compartment. The participation of the MAPK network in fundamental physiological processes, such as cell proliferation and inflammation, and the derangement of the homeostasis that occurs in disease processes, renders MAPK a highly desirable target for therapeutic intervention. As we enhance our comprehension of scaffolds and other regulatory molecules, novel targets for drug design may be discovered that will afford selective and specific MAPK modulation.
Collapse
Affiliation(s)
- M D Brown
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
41
|
Kupzig S, Bouyoucef D, Cozier GE, Cullen PJ. Studying the spatial and temporal regulation of Ras GTPase-activating proteins. Methods Enzymol 2008; 407:64-82. [PMID: 16757315 DOI: 10.1016/s0076-6879(05)07007-2] [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: 05/10/2023]
Abstract
Two classes of proteins govern Ras activation. Guanine-nucleotide exchange factors (Ras GEFs) catalyze the activation of Ras by inducing the dissociation of GDP to allow association of the more abundant GTP, whereas GTPase-activating proteins (Ras GAPs), bind to the GTP-bound form and, by enhancing the intrinsic GTPase activity, catalyze Ras inactivation. A wide range of Ras GEFs and Ras GAPs have been identified from the various genome projects, and in a few instances, the mechanisms by which signals originating from activated receptors converge on specific GEFs and GAPs have been mapped. However, for most Ras GEFs and GAPs we have a poor understanding of their regulation. Here we focus on describing methods used to study the regulation of the GAP1 family of Ras GAPs. In particular, we emphasize how by combining biochemical, molecular, and imaging techniques, one can determine some of the complex array of mechanisms that have evolved to modulate the spatial and temporal dynamics of Ras regulation through these various Ras GAPs. By combining biochemical, molecular, and imaging techniques, we describe the visualization of the diverse and dynamic mechanisms through which stimulation of cell surface receptors leads to the regulation of these proteins. Thus, although each member of the GAP1 family performs the same basic biological function, that is, they function as Ras GAPs, each is designed to respond and decode signals from distinct second messenger pathways.
Collapse
Affiliation(s)
- Sabine Kupzig
- The Henry Wellcome Integrated Signalling Laboratories, Department of Biochemistry, School of Medical Sciences, University of Bristol, Bristol, United Kingdom
| | | | | | | |
Collapse
|
42
|
Roderick HL, Cook SJ. Ca2+ signalling checkpoints in cancer: remodelling Ca2+ for cancer cell proliferation and survival. Nat Rev Cancer 2008; 8:361-75. [PMID: 18432251 DOI: 10.1038/nrc2374] [Citation(s) in RCA: 553] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Increases in cytosolic free Ca2+ ([Ca2+]i) represent a ubiquitous signalling mechanism that controls a variety of cellular processes, including proliferation, metabolism and gene transcription, yet under certain conditions increases in intracellular Ca2+ are cytotoxic. Thus, in using Ca2+ as a messenger, cells walk a tightrope in which [Ca2+]i is strictly maintained within defined boundaries. To adhere to these boundaries and to sustain their modified phenotype, many cancer cells remodel the expression or activity of their Ca2+ signalling apparatus. Here, we review the role of Ca2+ in promoting cell proliferation and cell death, how these processes are remodelled in cancer and the opportunities this might provide for therapeutic intervention.
Collapse
Affiliation(s)
- H Llewelyn Roderick
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK.
| | | |
Collapse
|
43
|
The C2 domain of SynGAP is essential for stimulation of the Rap GTPase reaction. EMBO Rep 2008; 9:350-5. [PMID: 18323856 DOI: 10.1038/embor.2008.20] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2007] [Revised: 01/16/2008] [Accepted: 01/17/2008] [Indexed: 11/08/2022] Open
Abstract
The brain-specific synaptic guanosine triphosphatase (GTPase)-activating protein (SynGAP) is important in synaptic plasticity. It shows dual specificity for the small guanine nucleotide-binding proteins Rap and Ras. Here, we show that RapGAP activity of SynGAP requires its C2 domain. In contrast to the isolated GAP domain, which does not show any detectable RapGAP activity, a fragment comprising the C2 and GAP domains (C2-GAP) stimulates the intrinsic GTPase reaction of Rap by approximately 1 x 10(4). The C2-GAP crystal structure, complemented by modelling and biochemical analyses, favours a concerted movement of the C2 domain towards the switch II region of Rap to assist in GTPase stimulation. Our data support a catalytic mechanism similar to that of canonical RasGAPs and distinct from the canonical RapGAPs. SynGAP presents the first example, to our knowledge, of a GAP that uses a second domain for catalytic activity, thus pointing to a new function of C2 domains.
Collapse
|
44
|
Evans JH, Falke JJ. Ca2+ influx is an essential component of the positive-feedback loop that maintains leading-edge structure and activity in macrophages. Proc Natl Acad Sci U S A 2007; 104:16176-81. [PMID: 17911247 PMCID: PMC2042181 DOI: 10.1073/pnas.0707719104] [Citation(s) in RCA: 123] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In migrating eukaryotic cells, phosphatidylinositol 3-kinase (PI3K), filamentous actin (F-actin), and monomeric Rho GTPases are key components of a complex positive-feedback system that maintains and amplifies a phosphatidylinositol-3,4,5-trisphosphate signal at the leading edge of the cell. This lipid signal is required for cell polarization and movement. In leukocytes and Dictyostelium, activation or inhibition of any one of these components leads to the activation or inhibition, respectively, of the others via undefined feedback interactions. The role of Ca(2+) signals in migrating leukocytes is controversial, and there has been no indication that Ca(2+) participates in positive feedback. Here, we demonstrate that an extracellular Ca(2+) influx is required for positive feedback at the leading edge of spontaneously polarized macrophages. Inhibition of extracellular Ca(2+) influx leads to loss of leading-edge PI3K activity, disassembly of F-actin, cessation of ruffling, and decay of chemoattractant signals. Conversely, increasing cytosolic Ca(2+) enhances membrane ruffling, PI3K activity, and F-actin accumulation. Overall, these findings demonstrate that an extracellular Ca(2+) influx is an essential component, together with PI3K and F-actin, of the positive-feedback cycle that maintains leading-edge structure and ruffling activity and that supports the chemoattractant response. Strikingly, the Ca(2+)-sensitive enzyme protein kinase Calpha (PKCalpha) is enriched at the leading edge, and its enrichment is sensitive to blockade of Ca(2+) influx, to inhibition of PI3K activity, and to F-actin depolymerization. These findings support the working hypothesis that a local, leading-edge Ca(2+) signal recruits PKCalpha as a central player in the positive-feedback loop.
Collapse
Affiliation(s)
- John H. Evans
- *Department of Chemistry and Biochemistry and
- To whom correspondence may be addressed. E-mail: or
| | - Joseph J. Falke
- *Department of Chemistry and Biochemistry and
- the Molecular Biophysics Program, University of Colorado, Boulder, CO 80309-0215
- To whom correspondence may be addressed. E-mail: or
| |
Collapse
|
45
|
|
46
|
Jin H, Wang X, Ying J, Wong AHY, Cui Y, Srivastava G, Shen ZY, Li EM, Zhang Q, Jin J, Kupzig S, Chan ATC, Cullen PJ, Tao Q. Epigenetic silencing of a Ca(2+)-regulated Ras GTPase-activating protein RASAL defines a new mechanism of Ras activation in human cancers. Proc Natl Acad Sci U S A 2007; 104:12353-8. [PMID: 17640920 PMCID: PMC1941473 DOI: 10.1073/pnas.0700153104] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Ras has achieved notoriety as an oncogene aberrantly activated in multiple human tumors. Approximately 30% of all human tumors express an oncogenic form of this GTPase that is locked in an active conformation as a result of being insensitive to Ras GTPase-activating proteins (GAPs), proteins that normally regulate the inactivation of Ras by enhancing its intrinsic GTPase activity. Besides oncogenic mutations in Ras, signaling by wild-type Ras is also frequently deregulated in tumors through aberrant coupling to activated cell surface receptors. This indicates that alternative mechanisms of aberrant wild-type Ras activation may be involved in tumorigenesis. Here, we describe another mechanism through which aberrant Ras activation is achieved in human cancers. We have established that Ras GTPase-activating-like protein (RASAL), a Ca(2+)-regulated Ras GAP that decodes the frequency of Ca(2+) oscillations, is silenced through CpG methylation in multiple tumors. With the finding that ectopic expression of catalytically active RASAL leads to growth inhibition of these tumor cells by Ras inactivation, we have provided evidence that epigenetically silencing of this Ras GAP represents a mechanism of aberrant Ras activation in certain cancers. Our demonstration that RASAL constitutes a tumor suppressor gene has therefore further emphasized the importance of Ca(2+) in the regulation of Ras signaling and has established that deregulation of this pathway is an important step in Ras-mediated tumorigenesis.
Collapse
Affiliation(s)
- Hongchuan Jin
- *Cancer Epigenetics Laboratory, State Key Laboratory in Oncology in South China, Sir Y. K. Pao Center for Cancer, Department of Clinical Oncology, Hong Kong Cancer Institute and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong
| | - Xian Wang
- *Cancer Epigenetics Laboratory, State Key Laboratory in Oncology in South China, Sir Y. K. Pao Center for Cancer, Department of Clinical Oncology, Hong Kong Cancer Institute and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong
| | - Jianming Ying
- *Cancer Epigenetics Laboratory, State Key Laboratory in Oncology in South China, Sir Y. K. Pao Center for Cancer, Department of Clinical Oncology, Hong Kong Cancer Institute and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong
| | - Ada H. Y. Wong
- *Cancer Epigenetics Laboratory, State Key Laboratory in Oncology in South China, Sir Y. K. Pao Center for Cancer, Department of Clinical Oncology, Hong Kong Cancer Institute and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong
| | - Yan Cui
- *Cancer Epigenetics Laboratory, State Key Laboratory in Oncology in South China, Sir Y. K. Pao Center for Cancer, Department of Clinical Oncology, Hong Kong Cancer Institute and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong
| | | | - Zhong-Ying Shen
- Shantou University Medical College/Chinese University of Hong Kong Joint Epigenetics Group, Shantou University Medical College, Shantou 515041, China
| | - En-Min Li
- Shantou University Medical College/Chinese University of Hong Kong Joint Epigenetics Group, Shantou University Medical College, Shantou 515041, China
| | - Qian Zhang
- Department of Urology, Peking University First Hospital and Institute of Urology, Beijing 100034, China; and
| | - Jie Jin
- Department of Urology, Peking University First Hospital and Institute of Urology, Beijing 100034, China; and
| | - Sabine Kupzig
- The Henry Wellcome Integrated Signalling Laboratories, Department of Biochemistry, School of Medical Sciences, University of Bristol, Bristol BS8 1TD, United Kingdom
| | - Anthony T. C. Chan
- *Cancer Epigenetics Laboratory, State Key Laboratory in Oncology in South China, Sir Y. K. Pao Center for Cancer, Department of Clinical Oncology, Hong Kong Cancer Institute and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong
| | - Peter J. Cullen
- The Henry Wellcome Integrated Signalling Laboratories, Department of Biochemistry, School of Medical Sciences, University of Bristol, Bristol BS8 1TD, United Kingdom
- To whom correspondence may be addressed. E-mail: or
| | - Qian Tao
- *Cancer Epigenetics Laboratory, State Key Laboratory in Oncology in South China, Sir Y. K. Pao Center for Cancer, Department of Clinical Oncology, Hong Kong Cancer Institute and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong
- Shantou University Medical College/Chinese University of Hong Kong Joint Epigenetics Group, Shantou University Medical College, Shantou 515041, China
- To whom correspondence may be addressed. E-mail: or
| |
Collapse
|
47
|
Yarwood S, Bouyoucef-Cherchalli D, Cullen PJ, Kupzig S. The GAP1 family of GTPase-activating proteins: spatial and temporal regulators of small GTPase signalling. Biochem Soc Trans 2007; 34:846-50. [PMID: 17052212 DOI: 10.1042/bst0340846] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Ras proteins are binary switches that, by cycling between inactive GDP-bound and active GTP-bound conformations, regulate multiple cellular signalling pathways including those that control cell growth, differentiation and survival. Approximately 30% of all human tumours express Ras-containing oncogenic mutations that lock the protein into a constitutively active conformation. The activation status of Ras is regulated by two groups of proteins: GEFs (guanine nucleotide-exchange factors) bind to Ras and enhance the exchange of GDP for GTP, thereby activating it, whereas GAPs (GTPase-activating proteins) inactivate Ras by binding to the GTP-bound form and enhancing the hydrolysis of the bound nucleotide back to GDP. In this review, we focus on a group of key regulators of Ras inactivation, the GAP1 family of Ras-GAPs. The members of this family are GAP1m, GAP1IP4BP, CAPRI (Ca2+-promoted Ras inactivator) and RASAL (Ras-GTPase-activating-like protein) and, as we will discuss, they are emerging as important modulators of Ras and small GTPase signalling that are subject to regulation by a diverse array of events and second messenger signals.
Collapse
Affiliation(s)
- S Yarwood
- The Henry Wellcome Integrated Signalling Laboratories, Department of Biochemistry, School of Medical Sciences, University of Bristol, Bristol BS8 1TD, UK
| | | | | | | |
Collapse
|
48
|
Rajalingam K, Schreck R, Rapp UR, Albert S. Ras oncogenes and their downstream targets. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2007; 1773:1177-95. [PMID: 17428555 DOI: 10.1016/j.bbamcr.2007.01.012] [Citation(s) in RCA: 318] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2006] [Revised: 01/17/2007] [Accepted: 01/19/2007] [Indexed: 12/30/2022]
Abstract
RAS proteins are small GTPases, which serve as master regulators of a myriad of signaling cascades involved in highly diverse cellular processes. RAS oncogenes have been originally discovered as retroviral oncogenes, and ever since constitutively activating RAS mutations have been identified in human tumors, they are in the focus of intense research. In this review, we summarize the biochemical properties of RAS proteins, trace down the evolution of RAS signaling and present an overview of the spatio-temporal activation of major RAS isoforms. We further discuss RAS effector pathways, their role in normal and transformed cell physiology and summarize ongoing attempts to interfere with aberrant RAS signaling. Finally, we comment on the role of micro RNAs in modulating RAS expression, contribution of RAS to stem cell function and on high-throughput analyses of RAS signaling networks.
Collapse
Affiliation(s)
- Krishnaraj Rajalingam
- University of Würzburg, Institut für Medizinische Strahlenkunde und Zellforschung, Versbacherstr. 5, D-97078 Würzburg, Germany
| | | | | | | |
Collapse
|
49
|
Brown DPG, Gökmen-Polar Y, Jiang L, Tan J, Ringham H, Janecki DJ, Qi G, Witzmann FA, Sledge GW, Wang M. A comparative proteomic study to characterize the vinblastine resistance in human ovarian cancer cells. Proteomics Clin Appl 2007; 1:18-31. [DOI: 10.1002/prca.200600171] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2006] [Indexed: 11/10/2022]
|
50
|
Felsani A, Mileo AM, Maresca V, Picardo M, Paggi MG. New technologies used in the study of human melanoma. INTERNATIONAL REVIEW OF CYTOLOGY 2007; 261:247-86. [PMID: 17560284 DOI: 10.1016/s0074-7696(07)61006-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The amount of information on tumor biology has expanded enormously, essentially due to the completion of the human genome sequencing and to the application of new technologies that represent an exciting breakthrough in molecular analysis. Often these data spring from experimental procedures, such as a serial analysis of gene expression (SAGE) and DNA microarrays, which cannot be defined as hypothesis-driven: it may appear to be a "brute force" approach through which no information can be directly generated concerning the specific functions of selected genes in a definite context. However, interesting results are fruitfully generated, and thus it is important to consider the enormous potential these new technologies possess and to learn how to apply this novel form of knowledge in the emerging field of molecular medicine. This review, after a limited outline regarding several classic aspects of human cutaneous melanoma biology, genetics, and clinical approaches, will focus on the proficient use of up-to-date technologies in the study of the neoplastic disease and on their capability to provide effective support to conventional approaches in melanoma diagnosis, prognosis, and treatment.
Collapse
Affiliation(s)
- Armando Felsani
- CNR, Istituto di Neurobiologia e Medicina Molecolare, 00143 Rome, Italy
| | | | | | | | | |
Collapse
|