1
|
Wurtzel JGT, Lazar S, Askari S, Zhao X, Severa J, Ayombil F, Michael JV, Camire RM, McKenzie SE, Stalker TJ, Ma P, Goldfinger LE. Plasma growth factors maintain constitutive translation in platelets to regulate reactivity and thrombotic potential. Blood Adv 2024; 8:1550-1566. [PMID: 38163324 PMCID: PMC10982986 DOI: 10.1182/bloodadvances.2023011734] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/14/2023] [Accepted: 11/30/2023] [Indexed: 01/03/2024] Open
Abstract
ABSTRACT Mechanisms of proteostasis in anucleate circulating platelets are unknown and may regulate platelet function. We investigated the hypothesis that plasma-borne growth factors/hormones (GFHs) maintain constitutive translation in circulating platelets to facilitate reactivity. Bio-orthogonal noncanonical amino acid tagging (BONCAT) coupled with liquid chromatography-tandem mass spectrometry analysis revealed constitutive translation of a broad-spectrum translatome in human platelets dependent upon plasma or GFH exposure, and in murine circulation. Freshly isolated platelets from plasma showed homeostatic activation of translation-initiation signaling pathways: phosphorylation of p38/ERK upstream kinases, essential intermediate MNK1/2, and effectors eIF4E/4E-BP1. Plasma starvation led to loss of pathway phosphorylation, but it was fully restored with 5-minute stimulation by plasma or GFHs. Cycloheximide or puromycin infusion suppressed ex vivo platelet GpIIb/IIIa activation and P-selectin exposure with low thrombin concentrations and low-to-saturating concentrations of adenosine 5'-diphosphate (ADP) or thromboxane analog but not convulxin. ADP-induced thromboxane generation was blunted by translation inhibition, and secondary-wave aggregation was inhibited in a thromboxane-dependent manner. Intravenously administered puromycin reduced injury-induced clot size in cremaster muscle arterioles, and delayed primary hemostasis after tail tip amputation but did not delay neither final hemostasis after subsequent rebleeds, nor final hemostasis after jugular vein puncture. In contrast, these mice were protected from injury-induced arterial thrombosis and thrombin-induced pulmonary thromboembolism (PE), and adoptive transfer of translation-inhibited platelets into untreated mice inhibited arterial thrombosis and PE. Thus, constitutive plasma GFH-driven translation regulates platelet G protein-coupled receptor reactivity to balance hemostasis and thrombotic potential.
Collapse
Affiliation(s)
- Jeremy G. T. Wurtzel
- Division of Hematology, Department of Medicine, Cardeza Foundation for Hematologic Research, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Sophia Lazar
- Division of Hematology, Department of Medicine, Cardeza Foundation for Hematologic Research, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Shayan Askari
- Division of Hematology, Department of Medicine, Cardeza Foundation for Hematologic Research, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Xuefei Zhao
- Division of Hematology, Department of Medicine, Cardeza Foundation for Hematologic Research, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Jenna Severa
- Division of Hematology, Department of Medicine, Cardeza Foundation for Hematologic Research, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Francis Ayombil
- Division of Hematology and the Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - James V. Michael
- Division of Hematology, Department of Medicine, Cardeza Foundation for Hematologic Research, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Rodney M. Camire
- Division of Hematology and the Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Steven E. McKenzie
- Division of Hematology, Department of Medicine, Cardeza Foundation for Hematologic Research, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Timothy J. Stalker
- Division of Hematology, Department of Medicine, Cardeza Foundation for Hematologic Research, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Peisong Ma
- Division of Hematology, Department of Medicine, Cardeza Foundation for Hematologic Research, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Lawrence E. Goldfinger
- Division of Hematology, Department of Medicine, Cardeza Foundation for Hematologic Research, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| |
Collapse
|
2
|
Lazar S, Wurtzel JGT, Chen X, Ma P, Goldfinger LE. High-efficiency unassisted transfection of platelets with naked double-stranded miRNAs modulates signal-activated translation and platelet function. Platelets 2020; 32:794-806. [PMID: 32838617 DOI: 10.1080/09537104.2020.1809642] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
We sought novel approaches to improve transfection efficiencies of microRNAs (miRNAs) in platelets, and to apply these approaches to investigate the roles of miRNAs in regulating signal-activated protein translation and functional effects. We found that ex vivo human platelets support gymnosis---internalization of ectopic miRNAs following co-incubation in the absence of conventional transfection reagents or schemes---and subsequently incorporate transfected miRNA into ARGONAUTE2 (AGO2)-based RNA-induced silencing complexes (RISC). Thrombin/fibrinogen stimulation activated translation of miR-223-3p target SEPTIN2, which was suppressed by miR-223-3p transfection in an AGO2/RISC-dependent manner. Thrombin/fibrinogen-induced exosome and microvesicle generation was inhibited by miR-223-3p transfection, and this effect was reversed with a RISC inhibitor. Platelet gymnosis of naked miRNAs appeared to be mediated in part by endocytic pathways including clathrin-dependent and fluid-phase endocytosis and caveolae. These results demonstrate the ability of ex vivo platelets to internalize ectopic miRNAs by unassisted transfection, and utilize them to modulate signal-activated translation and platelet function. Our results identify new roles for miR-223-3p in extracellular vesicle generation in stimulated platelets. High-efficiency gymnotic transfection of miRNAs in ex vivo platelets may be a broadly useful tool for exploring molecular genetic regulation of platelet function.
Collapse
Affiliation(s)
- Sophia Lazar
- Cardeza Foundation for Hematologic Research, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jeremy G T Wurtzel
- Cardeza Foundation for Hematologic Research, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Xi Chen
- Cardeza Foundation for Hematologic Research, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Peisong Ma
- Cardeza Foundation for Hematologic Research, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Lawrence E Goldfinger
- Cardeza Foundation for Hematologic Research, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
3
|
Cai G, Pan S, Feng N, Zou H, Gu J, Yuan Y, Liu X, Liu Z, Bian J. Zearalenone inhibits T cell chemotaxis by inhibiting cell adhesion and migration related proteins. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 175:263-271. [PMID: 30903882 DOI: 10.1016/j.ecoenv.2019.03.045] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/10/2019] [Accepted: 03/12/2019] [Indexed: 06/09/2023]
Abstract
Zearalenone (ZEA) is a phenolic resorcylic acid lactone mycotoxin produced by several Fusarium species that grow on temperate and tropical crops. The number of reports documenting the immunotoxic effects of ZEA is increasing, but the underlying mechanism is not clear. The purpose of this study was to investigate the effects of ZEA on T cell chemotaxis and evaluate changes in adhesion and migration proteins associated with this process. Specifically, T cells were isolated from BALB/C mouse splenic lymphocytes, activated by concanavalin A (Con A), and then exposed to different concentrations of ZEA. Transmission electron microscopy (TEM) and scanning electron microscopy (SEM) were used observe the ultrastructural changes inside the cell and on the cell surface, respectively. The transwell migration assay was used to evaluate the effect of ZEA on T cell chemotaxis in the presence of CCL19 or CCL21. A confocal 3D laser was used to capture the morphology of perforated cells and western blot was used to detect the expression of proteins associated with cell migration and adhesion. Additionally, we used flow cytometry to examine the expression of chemokine receptors on T cells. Finally, the chemokine (RANTES and MIP-1α) levels secreted by T cells were assessed using cytometric bead array. Overall, our data showed that treatment with ZEA caused ultrastructural damage on the surface as well as inside of T cells. Moreover, ZEA inhibited T cell chemotaxis which was mediated by CCL19 or CCL21 and disrupted the balance of T cell subtypes. The expression of T cell adhesion and migration proteins was also inhibited by ZEA. The expression of T cell chemokine receptor as well as secretion of RANTES and MIP-1α by T cells was suppressed after ZEA treatment. In summary, our results indicate that ZEA reduced the chemotactic effect of T cells mediated by chemokines, which was likely linked to the inhibition of T cell motility and accompanied by decreased expression of adhesion and migration proteins.
Collapse
Affiliation(s)
- Guodong Cai
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China; .Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, Jiangsu, China
| | - Shunye Pan
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China; China Animal Husbandry Group, Beijing, 100000, China
| | - Nannan Feng
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China; .Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, China
| | - Hui Zou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China; .Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, China
| | - Jianhong Gu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China; .Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, China
| | - Yan Yuan
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China; .Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, China
| | - Xuezhong Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China; .Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, China
| | - Zongping Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China; .Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, Jiangsu, China
| | - Jianchun Bian
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China; .Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, Jiangsu, China.
| |
Collapse
|
4
|
Defective RAB1B-related megakaryocytic ER-to-Golgi transport in RUNX1 haplodeficiency: impact on von Willebrand factor. Blood Adv 2019; 2:797-806. [PMID: 29632235 DOI: 10.1182/bloodadvances.2017014274] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 02/27/2018] [Indexed: 12/18/2022] Open
Abstract
Patients with RUNX1 haplodeficiency have thrombocytopenia, platelet dysfunction, and deficiencies of α-granules and dense granules. Platelet expression profiling of a patient with a heterozygous RUNX1 mutation (c.969-323G>T) revealed decreased RAB1B, which encodes a small G protein. RAB GTPases regulate vesicle trafficking, and RAB1B is implicated in endoplasmic reticulum (ER)-to-Golgi transport in nonhematopoietic cells, but its role in megakaryocytes (MK) is unknown. We addressed the hypothesis that RAB1B is a transcriptional target of RUNX1 and that RAB1B regulates ER-to-Golgi transport in MK cells. Chromatin immunoprecipitation studies and electrophoretic mobility shift assay using phorbol 12-myristate 13-acetate (PMA)-treated human erythroleukemia cells revealed RUNX1 binding to RAB1B promoter region RUNX1 consensus sites, and their mutation reduced the promoter activity. RAB1B promoter activity and protein expression were inhibited by RUNX1 siRNA and enhanced by RUNX1 overexpression. These indicate that RAB1B is a direct RUNX1 target, providing a mechanism for decreased RAB1B in patient platelets. Vesicle trafficking from ER to Golgi in PMA-treated human erythroleukemia cells was impaired along with Golgi disruption on siRNA downregulation of RUNX1 or RAB1B. The effects of RUNX1 knockdown were reversed by RAB1B reconstitution. Trafficking of von Willebrand factor (vWF), an α-granule MK synthesized protein, was impaired with RUNX1 or RAB1B downregulation and reconstituted by ectopic RAB1B expression. Platelet vWF was decreased in patients with RUNX1 mutations. Thus, ER-to-Golgi transport, an early critical step in protein trafficking to granules, is impaired in megakaryocytic cells on RUNX1 downregulation, secondary to decreased RAB1B expression. Impaired RAB1B mediated ER-to-Golgi transport contributes to platelet α-granule defects in RUNX1 haplodeficiency.
Collapse
|
5
|
Strassburger K, Kang E, Teleman AA. Drosophila ZDHHC8 palmitoylates scribble and Ras64B and controls growth and viability. PLoS One 2019; 14:e0198149. [PMID: 30735487 PMCID: PMC6368284 DOI: 10.1371/journal.pone.0198149] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 01/24/2019] [Indexed: 12/20/2022] Open
Abstract
Palmitoylation is an important posttranslational modification regulating diverse cellular functions. Consequently, aberrant palmitoylation can lead to diseases such as neuronal disorders or cancer. In humans there are roughly one hundred times more palmitoylated proteins than enzymes catalyzing palmitoylation (palmitoyltransferases). Therefore, it is an important challenge to establish the links between palmitoyltransferases and their targets. From publicly available data, we find that expression of human ZDHHC8 correlates significantly with cancer survival. To elucidate the organismal function of ZDHHC8, we study the Drosophila ortholog of hZDHHC8, CG34449/dZDHHC8. Knockdown of dZDHHC8 causes tissue overgrowth while dZDHHC8 mutants are larval lethal. We provide a list of 159 palmitoylated proteins in Drosophila and present data suggesting that scribble and Ras64B are targets of dZDHHC8.
Collapse
Affiliation(s)
- Katrin Strassburger
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg University, Heidelberg, Germany
| | - Evangeline Kang
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg University, Heidelberg, Germany
| | - Aurelio A. Teleman
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg University, Heidelberg, Germany
- * E-mail:
| |
Collapse
|
6
|
Rac1 Nanoscale Organization on the Plasma Membrane Is Driven by Lipid Binding Specificity Encoded in the Membrane Anchor. Mol Cell Biol 2018; 38:MCB.00186-18. [PMID: 29967243 DOI: 10.1128/mcb.00186-18] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 06/21/2018] [Indexed: 12/21/2022] Open
Abstract
Rac1 is a small guanine nucleotide binding protein that cycles between an inactive GDP-bound and active GTP-bound state to regulate cell motility and migration. Rac1 signaling is initiated from the plasma membrane (PM). Here, we used high-resolution spatial mapping and manipulation of PM lipid composition to define Rac1 nanoscale organization. We found that Rac1 proteins in the GTP- and GDP-bound states assemble into nonoverlapping nanoclusters; thus, Rac1 proteins undergo nucleotide-dependent segregation. Rac1 also selectively interacts with phosphatidic acid (PA) and phosphoinositol (3,4,5)-trisphosphate (PIP3), resulting in nanoclusters enriched in these lipids. These lipids are structurally important because depleting the PM of PA or PIP3 impairs both Rac1 PM binding and Rac1 nanoclustering. Lipid binding specificity of Rac1 is encoded in the amino acid sequence of the polybasic domain (PBD) of the C-terminal membrane anchor. Point mutations within the PBD, including arginine-to-lysine substitutions, profoundly alter Rac1 lipid binding specificity without changing the electrostatics of the protein and result in impaired macropinocytosis and decreased cell spreading. We propose that Rac1 nanoclusters act as lipid-based signaling platforms emulating the spatiotemporal organization of Ras proteins and show that the Rac1 PBD-prenyl anchor has a biological function that extends beyond simple electrostatic engagement with the PM.
Collapse
|
7
|
Truong D, Boddy KC, Canadien V, Brabant D, Fairn GD, D'Costa VM, Coyaud E, Raught B, Pérez-Sala D, Park WS, Heo WD, Grinstein S, Brumell JH. Salmonella
exploits host Rho GTPase signalling pathways through the phosphatase activity of SopB. Cell Microbiol 2018; 20:e12938. [DOI: 10.1111/cmi.12938] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 06/11/2018] [Accepted: 07/06/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Dorothy Truong
- Cell Biology Program; Hospital for Sick Children; Toronto ON Canada
- Department of Molecular Genetics; University of Toronto; Toronto ON Canada
| | - Kirsten C. Boddy
- Cell Biology Program; Hospital for Sick Children; Toronto ON Canada
- Institute of Medical Science; University of Toronto; Toronto ON Canada
| | | | - Danielle Brabant
- Cell Biology Program; Hospital for Sick Children; Toronto ON Canada
| | - Gregory D. Fairn
- Institute of Medical Science; University of Toronto; Toronto ON Canada
- Keenan Research Centre for Biomedical Science; St. Michael's Hospital; Toronto ON Canada
| | | | - Etienne Coyaud
- Princess Margaret Cancer Centre; University Health Network; Toronto Ontario Canada
| | - Brian Raught
- Princess Margaret Cancer Centre; University Health Network; Toronto Ontario Canada
- Department of Medical Biophysics; University of Toronto; Toronto Ontario Canada
| | - Dolores Pérez-Sala
- Department of Structural and Chemical Biology; Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas; Madrid Spain
| | - Wei Sun Park
- Department of Biological Sciences; Korea Advanced Institute of Science and Technology (KAIST); Daejeon Republic of Korea
| | - Won Do Heo
- Department of Biological Sciences; Korea Advanced Institute of Science and Technology (KAIST); Daejeon Republic of Korea
- Center for Cognition and Sociality; Institute of Basic Science (IBS); Daejeon Republic of Korea
| | - Sergio Grinstein
- Cell Biology Program; Hospital for Sick Children; Toronto ON Canada
- Institute of Medical Science; University of Toronto; Toronto ON Canada
- Keenan Research Centre for Biomedical Science; St. Michael's Hospital; Toronto ON Canada
- Department of Biochemistry; University of Toronto; Toronto ON Canada
| | - John H. Brumell
- Cell Biology Program; Hospital for Sick Children; Toronto ON Canada
- Department of Molecular Genetics; University of Toronto; Toronto ON Canada
- Institute of Medical Science; University of Toronto; Toronto ON Canada
- Sickkids IBD Centre; Hospital for Sick Children; Toronto ON Canada
| |
Collapse
|
8
|
Small GTPase RAS in multiple sclerosis - exploring the role of RAS GTPase in the etiology of multiple sclerosis. Small GTPases 2018; 11:312-319. [PMID: 30043672 DOI: 10.1080/21541248.2018.1502591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
RAS signaling is involved in the development of autoimmunity in general. Multiple sclerosis (MS) is a T cell-mediated autoimmune disease of the central nervous system. It is widely recognized that a reduction of Foxp3+ regulatory T (Treg) cells is an immunological hallmark of MS, but the underlying mechanisms are unclear. In experimental autoimmune models, N-Ras and K-Ras inhibition triggers an anti-inflammatory effect up-regulating, via foxp3 elevation, the numbers and the functional suppressive properties of Tregs. Similarly, an increase in natural Tregs number during Experimental Autoimmune Encephalomyelitis (EAE) in R-RAS -/- mice results in attenuated disease. In humans, only KRAS GTPase isoform is involved in mechanism causing tolerance defects in rheumatoid arthritis (RA). T cells from these patients have increased transcription of KRAS (but not NRAS). RAS genes are major drivers in human cancers. Consequently, there has been considerable interest in developing anti-RAS inhibitors for cancer treatment. Despite efforts, no anti-RAS therapy has succeeded in the clinic. The major strategy that has so far reached the clinic aimed to inhibit activated Ras indirectly through blocking its post-translational modification and inducing its mis-localization. The disappointing clinical outcome of Farnesyl Transferase Inhibitors (FTIs) in cancers has decreased interest in these drugs. However, FTIs suppress EAE by downregulation of myelin-reactive activated T-lymphocytes and statins are currently studied in clinical trials for MS. However, no pharmacologic approaches to targeting Ras proteins directly have yet succeeded. The therapeutic strategy to recover immune function through the restoration of impaired Tregs function with the mounting evidences regarding KRAS in autoimmune mediated disorder (MS, SLE, RA, T1D) suggest as working hypothesis the direct targeting KRAS activation using cancer-derived small molecules may be clinically relevant. ABBREVIATIONS FTIs: Farnesyl Transferase Inhibitors; MS: Multiple Sclerosis; RRMS: Relapsing Remitting Multiple Sclerosis; PPMS: Primary Progressive Multiple Sclerosis; Tregs: regulatory T-cells; Foxp3: Forkhead box P3; EAE: Experimental Autoimmune Encephalomyelitis; T1D: Type 1 Diabete; SLE: Systemic Lupus Erythematosus; RA: Rheumatoid Arthritis; CNS: Central Nervous System; TMEV: Theiler's murine encephalomyelitis virus; FTS: farnesyl thiosalicylic acid; TCR: T-Cell Receptor; AIA: Adjuvant-induced Arthritis; EAN: experimental autoimmune neuritis; HVR: hypervariable region; HMG-CoA: 3-hydroxy-3-methylglutaryl coenzyme A reductase; PBMC: Peripheral Blood Mononuclear Cells.
Collapse
|
9
|
Concepts and advances in cancer therapeutic vulnerabilities in RAS membrane targeting. Semin Cancer Biol 2017; 54:121-130. [PMID: 29203271 DOI: 10.1016/j.semcancer.2017.11.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 11/30/2017] [Indexed: 01/05/2023]
Abstract
For decades oncogenic RAS proteins were considered undruggable due to a lack of accessible binding pockets on the protein surfaces. Seminal early research in RAS biology uncovered the basic paradigm of post-translational isoprenylation of RAS polypeptides, typically with covalent attachment of a farnesyl group, leading to isoprenyl-mediated RAS anchorage at the plasma membrane and signal initiation at those sites. However, the failure of farnesyltransferase inhibitors to translate to the clinic stymied anti-RAS therapy development. Over the past ten years, a more complete picture has emerged of RAS protein maturation, intracellular trafficking, and location, positioning and retention in subdomains at the plasma membrane, with a corresponding expansion in our understanding of how these properties of RAS contribute to signal outputs. Each of these aspects of RAS regulation presents a potential vulnerability in RAS function that may be exploited for therapeutic targeting, and inhibitors have been identified or developed that interfere with RAS for nearly all of them. This review will summarize current understanding of RAS membrane targeting with a focus on highlighting development and outcomes of inhibitors at each step.
Collapse
|
10
|
Platelet microparticles infiltrating solid tumors transfer miRNAs that suppress tumor growth. Blood 2017; 130:567-580. [PMID: 28500171 DOI: 10.1182/blood-2016-11-751099] [Citation(s) in RCA: 183] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 05/08/2017] [Indexed: 12/13/2022] Open
Abstract
Platelet-derived microparticles (PMPs) are associated with enhancement of metastasis and poor cancer outcomes. Circulating PMPs transfer platelet microRNAs (miRNAs) to vascular cells. Solid tumor vasculature is highly permeable, allowing the possibility of PMP-tumor cell interaction. Here, we show that PMPs infiltrate solid tumors in humans and mice and transfer platelet-derived RNA, including miRNAs, to tumor cells in vivo and in vitro, resulting in tumor cell apoptosis. MiR-24 was a major species in this transfer. PMP transfusion inhibited growth of both lung and colon carcinoma ectopic tumors, whereas blockade of miR-24 in tumor cells accelerated tumor growth in vivo, and prevented tumor growth inhibition by PMPs. Conversely, Par4-deleted mice, which had reduced circulating microparticles (MPs), supported accelerated tumor growth which was halted by PMP transfusion. PMP targeting was associated with tumor cell apoptosis in vivo. We identified direct RNA targets of platelet-derived miR-24 in tumor cells, which included mitochondrial mt-Nd2, and Snora75, a noncoding small nucleolar RNA. These RNAs were suppressed in PMP-treated tumor cells, resulting in mitochondrial dysfunction and growth inhibition, in an miR-24-dependent manner. Thus, platelet-derived miRNAs transfer in vivo to tumor cells in solid tumors via infiltrating MPs, regulate tumor cell gene expression, and modulate tumor progression. These findings provide novel insight into mechanisms of horizontal RNA transfer and add multiple layers to the regulatory roles of miRNAs and PMPs in tumor progression. Plasma MP-mediated transfer of regulatory RNAs and modulation of gene expression may be a common feature with important outcomes in contexts of enhanced vascular permeability.
Collapse
|
11
|
Kummola L, Ortutay Z, Vähätupa M, Prince S, Uusitalo-Järvinen H, Järvinen TAH, Junttila IS. R-Ras deficiency does not affect papain-induced IgE production in mice. IMMUNITY INFLAMMATION AND DISEASE 2017; 5:280-288. [PMID: 28497586 PMCID: PMC5569372 DOI: 10.1002/iid3.168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 03/29/2017] [Accepted: 04/13/2017] [Indexed: 01/12/2023]
Abstract
Introduction R‐Ras GTPase has recently been implicated in the regulation of immune functions, particularly in dendritic cell (DC) maturation, immune synapse formation, and subsequent T cell responses. Methods Here, we investigated the role of R‐Ras in allergen‐induced immune response (type 2 immune response) in Rras deficient (R‐Ras KO) and wild type (WT) mice. Results Initially, we found that the number of conventional DC's in the lymph nodes (LNs) was reduced in R‐Ras KO mice. The expression of co‐stimulatory CD80 and CD86 molecules on these cells was also reduced on DC's from the R‐Ras KO mice. However, there was no difference in papain‐induced immune response between the R‐Ras WT and KO as measured by serum IgE levels after the immunization. Interestingly, neither the DC number nor co‐stimulatory molecule expression was different between WT and R‐Ras KO animals after the immunization. Conclusions Taken together, despite having reduced number of conventional DC's in the R‐Ras KO mice and low expression of CD80 on DC's, the R‐Ras KO mice are capable of mounting papain‐induced IgE responses comparable to that of the WT mice. To our knowledge, this is the first report addressing potential differences in in vivo allergen responses regulated by the R‐Ras GTPase.
Collapse
Affiliation(s)
- Laura Kummola
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Zsuzsanna Ortutay
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Maria Vähätupa
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Stuart Prince
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Hannele Uusitalo-Järvinen
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland.,Departments of Ophthalmology and Orthopaedics & Traumatology, Tampere University Hospital, Tampere, Finland
| | - Tero A H Järvinen
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland.,Departments of Ophthalmology and Orthopaedics & Traumatology, Tampere University Hospital, Tampere, Finland
| | - Ilkka S Junttila
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland.,Fimlab Laboratories, Tampere, Finland
| |
Collapse
|
12
|
Goldfinger LE, Michael JV. Regulation of Ras signaling and function by plasma membrane microdomains. Biosci Trends 2017; 11:23-40. [PMID: 28179601 DOI: 10.5582/bst.2016.01220] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Together H-, N- and KRAS mutations are major contributors to ~30% of all human cancers. Thus, Ras inhibition remains an important anti-cancer strategy. The molecular mechanisms of isotypic Ras oncogenesis are still not completely understood. Monopharmacological therapeutics have not been successful in the clinic. These disappointing outcomes have led to attempts to target elements downstream of Ras, mainly targeting either the Phosphatidylinositol 3-Kinase (PI3K) or Mitogen-Activated Protein Kinase (MAPK) pathways. While several such approaches are moderately effective, recent efforts have focused on preclinical evaluation of combination therapies to improve efficacies. This review will detail current understanding of the contributions of plasma membrane microdomain targeting of Ras to mitogenic and tumorigenic signaling and tumor progression. Moreover, this review will outline novel approaches to target Ras in cancers, including targeting schemes for new drug development, as well as putative re-purposing of drugs in current use to take advantage of blunting Ras signaling by interfering with Ras plasma membrane microdomain targeting and retention.
Collapse
Affiliation(s)
- Lawrence E Goldfinger
- Department of Anatomy & Cell Biology and The Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine at Temple University, and Cancer Biology Program, Fox Chase Cancer Center
| | | |
Collapse
|
13
|
Regulation of H-Ras-driven MAPK signaling, transformation and tumorigenesis, but not PI3K signaling and tumor progression, by plasma membrane microdomains. Oncogenesis 2016; 5:e228. [PMID: 27239960 PMCID: PMC4945753 DOI: 10.1038/oncsis.2016.36] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 04/17/2016] [Indexed: 12/16/2022] Open
Abstract
In this study, we assessed the contributions of plasma membrane (PM) microdomain targeting to the functions of H-Ras and R-Ras. These paralogs have identical effector-binding regions, but variant C-terminal targeting domains (tDs) which are responsible for lateral microdomain distribution: activated H-Ras targets to lipid ordered/disordered (Lo/Ld) domain borders, and R-Ras to Lo domains (rafts). We hypothesized that PM distribution regulates Ras-effector interactions and downstream signaling. We used tD swap mutants, and assessed effects on signal transduction, cell proliferation, transformation and tumorigenesis. R-Ras harboring the H-Ras tD (R-Ras-tH) interacted with Raf, and induced Raf and ERK phosphorylation similar to H-Ras. R-Ras-tH stimulated proliferation and transformation in vitro, and these effects were blocked by both MEK and PI3K inhibition. Conversely, the R-Ras tD suppressed H-Ras-mediated Raf activation and ERK phosphorylation, proliferation and transformation. Thus, Ras access to Raf at the PM is sufficient for MAPK activation and is a principal component of Ras mitogenesis and transformation. Fusion of the R-Ras extended N-terminal domain to H-Ras had no effect on proliferation, but inhibited transformation and tumor progression, indicating that the R-Ras N-terminus also contributes negative regulation to these Ras functions. PI3K activation was tD independent; however, H-Ras was a stronger activator of PI3K than R-Ras, with either tD. PI3K inhibition nearly ablated transformation by R-Ras-tH, H-Ras and H-Ras-tR, whereas MEK inhibition had a modest effect on Ras-tH-driven transformation but no effect on H-Ras-tR transformation. R-Ras-tH supported tumor initiation, but not tumor progression. While H-Ras-tR-induced transformation was reduced relative to H-Ras, tumor progression was robust and similar to H-Ras. H-Ras tumor growth was moderately suppressed by MEK inhibition, which had no effect on H-Ras-tR tumor growth. In contrast, PI3K inhibition markedly suppressed tumor growth by H-Ras and H-Ras-tR, indicating that sustained PI3K signaling is a critical pathway for H-Ras-driven tumor progression, independent of microdomains.
Collapse
|
14
|
Control of Dendritic Spine Morphological and Functional Plasticity by Small GTPases. Neural Plast 2016; 2016:3025948. [PMID: 26989514 PMCID: PMC4775798 DOI: 10.1155/2016/3025948] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 01/06/2016] [Accepted: 01/19/2016] [Indexed: 11/18/2022] Open
Abstract
Structural plasticity of excitatory synapses is a vital component of neuronal development, synaptic plasticity, and behaviour. Abnormal development or regulation of excitatory synapses has also been strongly implicated in many neurodevelopmental, psychiatric, and neurodegenerative disorders. In the mammalian forebrain, the majority of excitatory synapses are located on dendritic spines, specialized dendritic protrusions that are enriched in actin. Research over recent years has begun to unravel the complexities involved in the regulation of dendritic spine structure. The small GTPase family of proteins have emerged as key regulators of structural plasticity, linking extracellular signals with the modulation of dendritic spines, which potentially underlies their ability to influence cognition. Here we review a number of studies that examine how small GTPases are activated and regulated in neurons and furthermore how they can impact actin dynamics, and thus dendritic spine morphology. Elucidating this signalling process is critical for furthering our understanding of the basic mechanisms by which information is encoded in neural circuits but may also provide insight into novel targets for the development of effective therapies to treat cognitive dysfunction seen in a range of neurological disorders.
Collapse
|
15
|
Yan X, Yan M, Guo Y, Singh G, Chen Y, Yu M, Wang D, Hillery CA, Chan AM. R-Ras Regulates Murine T Cell Migration and Intercellular Adhesion Molecule-1 Binding. PLoS One 2015; 10:e0145218. [PMID: 26710069 PMCID: PMC4692399 DOI: 10.1371/journal.pone.0145218] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Accepted: 11/30/2015] [Indexed: 12/04/2022] Open
Abstract
The trafficking of T-lymphocytes to peripheral draining lymph nodes is crucial for mounting an adaptive immune response. The role of chemokines in the activation of integrins via Ras-related small GTPases has been well established. R-Ras is a member of the Ras-subfamily of small guanosine-5’-triphosphate-binding proteins and its role in T cell trafficking has been investigated in R-Ras null mice (Rras−/−). An examination of the lymphoid organs of Rras−/− mice revealed a 40% reduction in the cellularity of the peripheral lymph nodes. Morphologically, the high endothelial venules of Rras−/− mice were more disorganized and less mature than those of wild-type mice. Furthermore, CD4+ and CD8+ T cells from Rras−/− mice had approximately 42% lower surface expression of L-selectin/CD62L. These aberrant peripheral lymph node phenotypes were associated with proliferative and trafficking defects in Rras−/− T cells. Furthermore, R-Ras could be activated by the chemokine, CCL21. Indeed, Rras−/− T cells had approximately 14.5% attenuation in binding to intercellular adhesion molecule 1 upon CCL21 stimulation. Finally, in a graft-versus host disease model, recipient mice that were transfused with Rras−/− T cells showed a significant reduction in disease severity when compared with mice transplanted with wild-type T cells. These findings implicate a role for R-Ras in T cell trafficking in the high endothelial venules during an effective immune response.
Collapse
Affiliation(s)
- Xiaocai Yan
- Department of Pediatrics, The Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Mingfei Yan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR
| | - Yihe Guo
- Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Gobind Singh
- Department of Oncological Sciences, The Mount Sinai School of Medicine, New York, New York, United States of America
| | - Yuhong Chen
- Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Mei Yu
- Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Demin Wang
- Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Cheryl A Hillery
- Department of Pediatrics, The Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America.,Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Andrew M Chan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR
| |
Collapse
|
16
|
RLIP76 regulates Arf6-dependent cell spreading and migration by linking ARNO with activated R-Ras at recycling endosomes. Biochem Biophys Res Commun 2015; 467:785-91. [PMID: 26498519 DOI: 10.1016/j.bbrc.2015.10.064] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 10/12/2015] [Indexed: 11/23/2022]
Abstract
R-Ras small GTPase enhances cell spreading and motility via RalBP1/RLIP76, an R-Ras effector that links GTP-R-Ras to activation of Arf6 and Rac1 GTPases. Here, we report that RLIP76 performs these functions by binding cytohesin-2/ARNO, an Arf GTPase guanine exchange factor, and connecting it to R-Ras at recycling endosomes. RLIP76 formed a complex with R-Ras and ARNO by binding ARNO via its N-terminus (residues 1-180) and R-Ras via residues 180-192. This complex was present in Rab11-positive recycling endosomes and the presence of ARNO in recycling endosomes required RLIP76, and was not supported by RLIP76(Δ1-180) or RLIP76(Δ180-192). Spreading and migration required RLIP76(1-180), and RLIP76(Δ1-180) blocked ARNO recruitment to recycling endosomes, and spreading. Arf6 activation with an ArfGAP inhibitor overcame the spreading defects in RLIP76-depleted cells or cells expressing RLIP76(Δ1-180). Similarly, RLIP76(Δ1-180) or RLIP76(Δ180-192) suppressed Arf6 activation. Together these results demonstrate that RLIP76 acts as a scaffold at recycling endosomes by binding activated R-Ras, recruiting ARNO to activate Arf6, thereby contributing to cell spreading and migration.
Collapse
|
17
|
Salem JC, Reviriego-Mendoza MM, Santy LC. ARF-GEF cytohesin-2/ARNO regulates R-Ras and α5-integrin recycling through an EHD1-positive compartment. Mol Biol Cell 2015; 26:4265-79. [PMID: 26378252 PMCID: PMC4642859 DOI: 10.1091/mbc.e15-05-0278] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 09/11/2015] [Indexed: 11/30/2022] Open
Abstract
R-Ras and cytohesin-2/ARNO coordinate in the control of epithelial cell adhesion, but the mechanism has been unclear. Cytohesin-2/ARNO regulates traffic through an EHD1-positive recycling compartment. Inhibition of cytohesin-2/ARNO activity traps R-Ras and integrins within the EHD1 compartment and impairs adhesion and spreading. When expressed in epithelial cells, cytohesin-2/ARNO, a guanine nucleotide exchange factor (GEF) for ARF small GTPases, causes a robust migration response. Recent evidence suggests that cytohesin-2/ARNO acts downstream of small the GTPase R-Ras to promote spreading and migration. We hypothesized that cytohesin-2/ARNO could transmit R-Ras signals by regulating the recycling of R-Ras through ARF activation. We found that Eps15-homology domain 1 (EHD1), a protein that associates with the endocytic recycling compartment (ERC), colocalizes with active R-Ras in transiently expressed HeLa cells. In addition, we show that EHD1-positive recycling endosomes are a novel compartment for cytohesin-2/ARNO. Knockdown or expression of GEF-inactive (E156K) cytohesin-2/ARNO causes R-Ras to accumulate on recycling endosomes containing EHD1 and inhibits cell spreading. E156K-ARNO also causes a reduction in focal adhesion size and number. Finally, we demonstrate that R-Ras/ARNO signaling is required for recycling of α5-integrin and R-Ras to the plasma membrane. These data establish a role for cytohesin-2/ARNO as a regulator of R-Ras and integrin recycling and suggest that ARF-regulated trafficking of R-Ras is required for R-Ras–dependent effects on spreading and adhesion formation.
Collapse
Affiliation(s)
- Joseph C Salem
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802
| | - Marta M Reviriego-Mendoza
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802
| | - Lorraine C Santy
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802
| |
Collapse
|
18
|
Blanc M, David F, Abrami L, Migliozzi D, Armand F, Bürgi J, van der Goot FG. SwissPalm: Protein Palmitoylation database. F1000Res 2015; 4:261. [PMID: 26339475 PMCID: PMC4544385 DOI: 10.12688/f1000research.6464.1] [Citation(s) in RCA: 203] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/06/2015] [Indexed: 12/19/2022] Open
Abstract
Protein S-palmitoylation is a reversible post-translational modification that regulates many key biological processes, although the full extent and functions of protein S-palmitoylation remain largely unexplored. Recent developments of new chemical methods have allowed the establishment of palmitoyl-proteomes of a variety of cell lines and tissues from different species. As the amount of information generated by these high-throughput studies is increasing, the field requires centralization and comparison of this information. Here we present SwissPalm (
http://swisspalm.epfl.ch), our open, comprehensive, manually curated resource to study protein S-palmitoylation. It currently encompasses more than 5000 S-palmitoylated protein hits from seven species, and contains more than 500 specific sites of S-palmitoylation. SwissPalm also provides curated information and filters that increase the confidence in true positive hits, and integrates predictions of S-palmitoylated cysteine scores, orthologs and isoform multiple alignments. Systems analysis of the palmitoyl-proteome screens indicate that 10% or more of the human proteome is susceptible to S-palmitoylation. Moreover, ontology and pathway analyses of the human palmitoyl-proteome reveal that key biological functions involve this reversible lipid modification. Comparative analysis finally shows a strong crosstalk between S-palmitoylation and other post-translational modifications. Through the compilation of data and continuous updates, SwissPalm will provide a powerful tool to unravel the global importance of protein S-palmitoylation.
Collapse
Affiliation(s)
- Mathieu Blanc
- Global Health Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Fabrice David
- Global Health Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland.,Bioinformatics and biostatistics Core Facility, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Laurence Abrami
- Global Health Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Daniel Migliozzi
- Global Health Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Florence Armand
- Proteomic Core Facility, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Jérôme Bürgi
- Global Health Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Françoise Gisou van der Goot
- Global Health Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| |
Collapse
|
19
|
Lee S, Wurtzel JGT, Goldfinger LE. The RLIP76 N-terminus binds ARNO to regulate PI 3-kinase, Arf6 and Rac signaling, cell spreading and migration. Biochem Biophys Res Commun 2014; 454:560-5. [PMID: 25450693 DOI: 10.1016/j.bbrc.2014.10.114] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 10/24/2014] [Indexed: 11/29/2022]
Abstract
RLIP76 is a multifunctional protein involved in tumor growth and angiogenesis, and a promising therapeutic target in many cancers. RLIP76 harbors docking sites for many proteins, and we have found that it interacts with ARNO, a guanine nucleotide exchange factor for Arf6, and that RLIP76 regulates activation of Rac1 via Arf6, and regulates cell spreading and migration in an ARNO and Arf6-dependent manner. Here we show that ARNO interacts with the RLIP76 N-terminal domain, and this domain was required for RLIP76-dependent cell spreading and migration. We identified two sites in the RLIP76 N-terminus with differential effects on ARNO binding and downstream signaling: Ser29/Ser30 and Ser62. Ser29/30 mutation to Alanine inhibited ARNO interaction and was sufficient to block RLIP76-dependent cell spreading and migration, as well as RLIP76-dependent Arf6 activation. In contrast, RLIP76(S62A) interacted with ARNO and supported Arf6 activation. However, both sets of mutations blocked Rac1 activation. RLIP76-mediated Rac and Arf6 activation required PI3K activity. S29/30A mutations inhibited RLIP76-dependent PI3K activation, but S62A mutation did not. Together these results show that ARNO interaction with the RLIP76 N-terminus regulates cell spreading and motility via PI3K and Arf6, independent of RLIP76 control of Rac.
Collapse
Affiliation(s)
- Seunghyung Lee
- Department of Anatomy & Cell Biology and The Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Jeremy G T Wurtzel
- Department of Anatomy & Cell Biology and The Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Lawrence E Goldfinger
- Department of Anatomy & Cell Biology and The Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA 19140, USA; Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.
| |
Collapse
|
20
|
Lee S, Goldfinger LE. RLIP76 regulates HIF-1 activity, VEGF expression and secretion in tumor cells, and secretome transactivation of endothelial cells. FASEB J 2014; 28:4158-68. [PMID: 24928198 DOI: 10.1096/fj.14-255711] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 06/02/2014] [Indexed: 02/02/2023]
Abstract
This study was undertaken to reveal the mechanisms by which RLIP76 regulates endothelial cell angiogenic responses. RLIP76 is an effector of the angiogenic modulator, R-Ras. RLIP76 is overexpressed in many tumors, required for tumor angiogenesis, and blockade of RLIP76 results in tumor regression in multiple models. We report here that RLIP76 was required for expression and secretion of vascular endothelial growth factor (VEGF) in carcinoma and melanoma cells. Conditioned medium derived from RLIP76-depleted tumor cells, but not control knockdown cells, could not stimulate proliferation, migration, or Matrigel cord formation in endothelial cell cultures, which indicates that RLIP76 regulates angiogenic components of the tumor cell secretome. Recombinant VEGF added to conditioned medium from RLIP76-knockdown tumor cells restored these endothelial cell functions. Transcriptional activity of hypoxia-inducible factor 1 (HIF-1), which drives VEGF expression, was blocked in RLIP76-depleted tumor cells. RLIP76 was required for PI3-kinase activation, known to regulate HIF-1, in these cells. However, HIF-1α expression and nuclear localization were unaffected by RLIP76 knockdown, which suggests that RLIP76 regulates HIF-1 at the functional level. Thus, RLIP76 regulates tumor cell transactivation of endothelial cells via control of VEGF expression and secretion, providing a new important link in the mechanism of tumor cell induction of angiogenesis.
Collapse
Affiliation(s)
- Seunghyung Lee
- Department of Anatomy and Cell Biology, Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, USA; and
| | - Lawrence E Goldfinger
- Department of Anatomy and Cell Biology, Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, USA; and Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| |
Collapse
|
21
|
Ray A, Basu S, Miller NM, Chan AM, Dittel BN. An increase in tolerogenic dendritic cell and natural regulatory T cell numbers during experimental autoimmune encephalomyelitis in Rras-/- mice results in attenuated disease. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 192:5109-17. [PMID: 24771856 PMCID: PMC4041102 DOI: 10.4049/jimmunol.1302254] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
R-Ras is a member of the Ras superfamily of small GTPases, which are regulators of various cellular processes, including adhesion, survival, proliferation, trafficking, and cytokine production. R-Ras is expressed by immune cells and has been shown to modulate dendritic cell (DC) function in vitro and has been associated with liver autoimmunity. We used Rras-deficient mice to study the mechanism whereby R-Ras contributes to autoimmunity using experimental autoimmune encephalomyelitis (EAE), a mouse model of the CNS autoimmune disease multiple sclerosis. We found that a lack of R-Ras in peripheral immune cells resulted in attenuated EAE disease. Further investigation revealed that, during EAE, absence of R-Ras promoted the formation of MHC II(low) DC concomitant with a significant increase in proliferation of natural regulatory T cells, resulting in an increase in their cell numbers in the periphery. Our study suggests a novel role for R-Ras in promoting autoimmunity through negative regulation of natural regulatory T cell numbers by inhibiting the development of MHCII(low) DC with tolerogenic potential.
Collapse
Affiliation(s)
- Avijit Ray
- Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI 53201
| | - Sreemanti Basu
- Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI 53201; Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI 53226; and
| | - Nichole M Miller
- Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI 53201
| | - Andrew M Chan
- Division of Hematology and Oncology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Bonnie N Dittel
- Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI 53201; Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI 53226; and
| |
Collapse
|
22
|
Silver L, Michael JV, Goldfinger LE, Gallo G. Activation of PI3K and R-Ras signaling promotes the extension of sensory axons on inhibitory chondroitin sulfate proteoglycans. Dev Neurobiol 2014; 74:918-33. [PMID: 24578264 DOI: 10.1002/dneu.22174] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 02/24/2014] [Indexed: 12/22/2022]
Abstract
Chondroitin sulfate proteoglycans (CSPGs) are extracellular inhibitors of axon extension and plasticity, and cause growth cones to exhibit dystrophic behaviors. Phosphoinositide 3-kinase (PI3K) is a lipid kinase activated by axon growth promoting signals. In this study, we used embryonic chicken dorsal root ganglion neurons to determine if CSPGs impair signaling through PI3K. We report that CSPGs inhibit PI3K signaling in axons and growth cones, as evidenced by decreased levels of phosphorylated downstream kinases (Akt and S6). Direct activation of PI3K signaling, using a cell permeable phosphopeptide (PI3Kpep), countered the effects of CSPGs on growth cones and axon extension. Both overnight and acute treatment with PI3Kpep promoted axon extension on CSPG-coated substrates. The R-Ras GTPase is an upstream positive regulator of PI3K signaling. Expression of constitutively active R-Ras promoted axon extension and growth cone elaboration on CSPGs and permissive substrata. In contrast, an N-terminus-deleted constitutively active R-Ras, deficient in PI3K activation, promoted axon extension but not growth cone elaboration on CSPGs and permissive substrata. These data indicate that activation of R-Ras-PI3K signaling may be a viable approach for manipulating axon extension on CSPGs.
Collapse
Affiliation(s)
- Lee Silver
- Department of Anatomy and Cell Biology, Temple University School of Medicine, 3500 North Broad St., Philadelphia, Pennsylvania, 19140; Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, 19140
| | | | | | | |
Collapse
|
23
|
Fujimoto M, Ueda T. Conserved and plant-unique mechanisms regulating plant post-Golgi traffic. FRONTIERS IN PLANT SCIENCE 2012; 3:197. [PMID: 22973281 PMCID: PMC3428585 DOI: 10.3389/fpls.2012.00197] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Accepted: 08/08/2012] [Indexed: 05/18/2023]
Abstract
Membrane traffic plays crucial roles in diverse aspects of cellular and organelle functions in eukaryotic cells. Molecular machineries regulating each step of membrane traffic including the formation, tethering, and fusion of membrane carriers are largely conserved among various organisms, which suggests that the framework of membrane traffic is commonly shared among eukaryotic lineages. However, in addition to the common components, each organism has also acquired lineage-specific regulatory molecules that may be associated with the lineage-specific diversification of membrane trafficking events. In plants, comparative genomic analyses also indicate that some key machineries of membrane traffic are significantly and specifically diversified. In this review, we summarize recent progress regarding plant-unique regulatory mechanisms for membrane traffic, with a special focus on vesicle formation and fusion components in the post-Golgi trafficking pathway.
Collapse
Affiliation(s)
- Masaru Fujimoto
- Department of Biological Sciences, Graduate School of Science, The University of TokyoTokyo, Japan
| | - Takashi Ueda
- Department of Biological Sciences, Graduate School of Science, The University of TokyoTokyo, Japan
- Japan Science and Technology Agency, Precursory Research for Embryonic Science and TechnologyKawaguchi, Japan
- *Correspondence: Takashi Ueda, Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan. e-mail:
| |
Collapse
|