1
|
Chen Y, Miyazono K, Otsuka Y, Kanamori M, Yamashita A, Arashiki N, Matsumoto T, Takada K, Sato K, Mohandas N, Inaba M. Membrane skeleton hyperstability due to a novel alternatively spliced 4.1R can account for ellipsoidal camelid red cells with decreased deformability. J Biol Chem 2023; 299:102877. [PMID: 36621628 PMCID: PMC9926112 DOI: 10.1016/j.jbc.2023.102877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 12/28/2022] [Accepted: 12/29/2022] [Indexed: 01/08/2023] Open
Abstract
The red blood cells (RBCs) of vertebrates have evolved into two basic shapes, with nucleated nonmammalian RBCs having a biconvex ellipsoidal shape and anuclear mammalian RBCs having a biconcave disk shape. In contrast, camelid RBCs are flat ellipsoids with reduced membrane deformability, suggesting altered membrane skeletal organization. However, the mechanisms responsible for their elliptocytic shape and reduced deformability have not been determined. We here showed that in alpaca RBCs, protein 4.1R, a major component of the membrane skeleton, contains an alternatively spliced exon 14-derived cassette (e14) not observed in the highly conserved 80 kDa 4.1R of other highly deformable biconcave mammalian RBCs. The inclusion of this exon, along with the preceding unordered proline- and glutamic acid-rich peptide (PE), results in a larger and unique 90 kDa camelid 4.1R. Human 4.1R containing e14 and PE, but not PE alone, showed markedly increased ability to form a spectrin-actin-4.1R ternary complex in viscosity assays. A similar facilitated ternary complex was formed by human 4.1R possessing a duplication of the spectrin-actin-binding domain, one of the mutations known to cause human hereditary elliptocytosis. The e14- and PE-containing mutant also exhibited an increased binding affinity to β-spectrin compared with WT 4.1R. Taken together, these findings indicate that 4.1R protein with the e14 cassette results in the formation and maintenance of a hyperstable membrane skeleton, resulting in rigid red ellipsoidal cells in camelid species, and suggest that membrane structure is evolutionarily regulated by alternative splicing of exons in the 4.1R gene.
Collapse
Affiliation(s)
- Yuqi Chen
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Kosuke Miyazono
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Yayoi Otsuka
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Mariko Kanamori
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Aozora Yamashita
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Nobuto Arashiki
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan; Department of Biochemistry, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Takehisa Matsumoto
- Drug Discovery Structural Biology Platform Unit, RIKEN Center for Biosystems Dynamics Research, Yokohama, Japan
| | - Kensuke Takada
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Kota Sato
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Narla Mohandas
- Red Cell Physiology Laboratory, New York Blood Center, New York, New York, USA
| | - Mutsumi Inaba
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan.
| |
Collapse
|
2
|
Huang Y, Hale J, Wang Y, Li W, Zhang S, Zhang J, Zhao H, Guo X, Liu J, Yan H, Yazdanbakhsh K, Huang G, Hillyer CD, Mohandas N, Chen L, Sun L, An X. SF3B1 deficiency impairs human erythropoiesis via activation of p53 pathway: implications for understanding of ineffective erythropoiesis in MDS. J Hematol Oncol 2018; 11:19. [PMID: 29433555 PMCID: PMC5810112 DOI: 10.1186/s13045-018-0558-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 01/23/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND SF3B1 is a core component of splicing machinery. Mutations in SF3B1 are frequently found in myelodysplastic syndromes (MDS), particularly in patients with refractory anemia with ringed sideroblasts (RARS), characterized by isolated anemia. SF3B1 mutations have been implicated in the pathophysiology of RARS; however, the physiological function of SF3B1 in erythropoiesis remains unknown. METHODS shRNA-mediated approach was used to knockdown SF3B1 in human CD34+ cells. The effects of SF3B1 knockdown on human erythroid cell differentiation, cell cycle, and apoptosis were assessed by flow cytometry. RNA-seq, qRT-PCR, and western blot analyses were used to define the mechanisms of phenotypes following knockdown of SF3B1. RESULTS We document that SF3B1 knockdown in human CD34+ cells leads to increased apoptosis and cell cycle arrest of early-stage erythroid cells and generation of abnormally nucleated late-stage erythroblasts. RNA-seq analysis of SF3B1-knockdown erythroid progenitor CFU-E cells revealed altered splicing of an E3 ligase Makorin Ring Finger Protein 1 (MKRN1) and subsequent activation of p53 pathway. Importantly, ectopic expression of MKRN1 rescued SF3B1-knockdown-induced alterations. Decreased expression of genes involved in mitosis/cytokinesis pathway including polo-like kinase 1 (PLK1) was noted in SF3B1-knockdown polychromatic and orthochromatic erythroblasts comparing to control cells. Pharmacologic inhibition of PLK1 also led to generation of abnormally nucleated erythroblasts. CONCLUSIONS These findings enabled us to identify novel roles for SF3B1 in human erythropoiesis and provided new insights into its role in regulating normal erythropoiesis. Furthermore, these findings have implications for improved understanding of ineffective erythropoiesis in MDS patients with SF3B1 mutations.
Collapse
Affiliation(s)
- Yumin Huang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000 Henan People’s Republic of China
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065 USA
| | - John Hale
- Red Cell Physiology Laboratory, New York Blood Center, New York, NY 10065 USA
| | - Yaomei Wang
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065 USA
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001 People’s Republic of China
| | - Wei Li
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065 USA
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001 People’s Republic of China
- Department of Immunology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008 People’s Republic of China
| | - Shijie Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001 People’s Republic of China
| | - Jieying Zhang
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065 USA
- The State Key Laboratory of Medical Genetics and School of Life Sciences, Central South University, Changsha, 410078 People’s Republic of China
| | - Huizhi Zhao
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001 People’s Republic of China
| | - Xinhua Guo
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065 USA
| | - Jing Liu
- The State Key Laboratory of Medical Genetics and School of Life Sciences, Central South University, Changsha, 410078 People’s Republic of China
| | - Hongxia Yan
- Red Cell Physiology Laboratory, New York Blood Center, New York, NY 10065 USA
| | - Karina Yazdanbakhsh
- Laboratory of Complement Biology, New York Blood Center, New York, NY 10065 USA
| | - Gang Huang
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229 USA
| | | | - Narla Mohandas
- Red Cell Physiology Laboratory, New York Blood Center, New York, NY 10065 USA
| | - Lixiang Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001 People’s Republic of China
| | - Ling Sun
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000 Henan People’s Republic of China
| | - Xiuli An
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000 Henan People’s Republic of China
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065 USA
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001 People’s Republic of China
| |
Collapse
|
3
|
Wu K, Zheng JL, Luo Z, Chen QL, Zhu QL, Wei-Hu. Carnitine palmitoyltransferase I gene in Synechogobius hasta: Cloning, mRNA expression and transcriptional regulation by insulin in vitro. Gene 2015; 576:429-40. [PMID: 26506441 DOI: 10.1016/j.gene.2015.10.055] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 10/20/2015] [Accepted: 10/22/2015] [Indexed: 11/28/2022]
Abstract
We cloned seven complete CPT I cDNA sequences (CPT I α1a-1a, CPT I α1a-1b, CPT I α1a-1c, CPT I α1a-2, CPT I α2a, CPT I α2b1a, CPT I β) and a partial cDNA sequence (CPT I α2b1b) from Synechogobius hasta. Phylogenetic analysis shows that there are four CPT I duplications in S. hasta, CPT I duplication resulting in CPT I α and CPT I β, CPT I α duplication producing CPT I α1 and CPT I α2, CPT I α2 duplication generating CPT I α2a and CPT I α2b, and CPT I α2b duplication creating CPT I α2b1a and CPT I α2b1b. Alternative splicing of CPT Iα1a results in the generation of four CPT I isoforms, CPT I α1a-1a, CPT I α1a-1b, CPT I α1a-1c and CPT I α1a-2. Five CPT I transcripts (CPT I α1a, CPT I α2a, CPT I α2b1a, CPT I α2b1b and CPT I β) mRNAs are expressed in a wide range of tissues, but their abundance of each CPT I mRNA shows the tissue-dependent expression patterns. Insulin incubation significantly reduces the mRNA expression of CPT Iα1a and CPT Iα2a, but not other transcripts in hepatocytes of S. hasta. For the first time, our study demonstrates CPT Iα2b duplication and CPT I α1a alternative splicing in fish at transcriptional level, and the CPT I mRNAs are differentially regulated by insulin in vitro, suggesting that four CPT I isoforms may play different physiological roles during insulin signaling.
Collapse
Affiliation(s)
- Kun Wu
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture of P.R.C., Fishery College, Huazhong Agricultural University, Wuhan 430070, China; Freshwater Aquaculture Collaborative Innovative Centre of Hubei Province, Wuhan 430070, China
| | - Jia-Lang Zheng
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture of P.R.C., Fishery College, Huazhong Agricultural University, Wuhan 430070, China; Freshwater Aquaculture Collaborative Innovative Centre of Hubei Province, Wuhan 430070, China
| | - Zhi Luo
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture of P.R.C., Fishery College, Huazhong Agricultural University, Wuhan 430070, China; Freshwater Aquaculture Collaborative Innovative Centre of Hubei Province, Wuhan 430070, China.
| | - Qi-Liang Chen
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture of P.R.C., Fishery College, Huazhong Agricultural University, Wuhan 430070, China; Freshwater Aquaculture Collaborative Innovative Centre of Hubei Province, Wuhan 430070, China
| | - Qing-Ling Zhu
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture of P.R.C., Fishery College, Huazhong Agricultural University, Wuhan 430070, China; Freshwater Aquaculture Collaborative Innovative Centre of Hubei Province, Wuhan 430070, China
| | - Wei-Hu
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture of P.R.C., Fishery College, Huazhong Agricultural University, Wuhan 430070, China; Freshwater Aquaculture Collaborative Innovative Centre of Hubei Province, Wuhan 430070, China
| |
Collapse
|
4
|
Satchwell TJ, Bell AJ, Toye AM. The sorting of blood group active proteins during enucleation. ISBT SCIENCE SERIES 2015; 10:163-168. [PMID: 26640516 PMCID: PMC4668593 DOI: 10.1111/voxs.12127] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Enucleation represents the critical stage during red blood cell development when the nucleus is extruded from an orthochromatic erythroblast in order to generate a nascent immature reticulocyte. Extrusion of the nucleus results in loss of a proportion of the erythroblast plasma membrane, which surrounds the nucleus, the bulk of the endoplasmic reticulum and a small region of cytoplasm. For this reason enucleation provides an important point in erythroblast differentiation at which proteins not required for the function of the erythrocyte can be lost, whilst those that are important for the structure-function properties of the mature erythrocyte must be efficiently retained in the reticulocyte plasma membrane. Disturbances in protein distribution during enucleation are envisaged to occur during human diseases such as Hereditary Spherocytosis. This article will discuss the current knowledge of erythroblast enucleation in the context of retention and loss of proteins that display antigenic blood group sites and that exist within multiprotein complexes within the erythrocyte membrane.
Collapse
Affiliation(s)
- Timothy J. Satchwell
- School of Biochemistry, Medical Sciences Building, University Walk, Bristol BS8 1TD
- Bristol Institute of Transfusion Sciences, NHS Blood and Transplant, Filton, Bristol
| | - Amanda J. Bell
- School of Biochemistry, Medical Sciences Building, University Walk, Bristol BS8 1TD
| | - Ashley M. Toye
- School of Biochemistry, Medical Sciences Building, University Walk, Bristol BS8 1TD
- Bristol Institute of Transfusion Sciences, NHS Blood and Transplant, Filton, Bristol
| |
Collapse
|
5
|
Bazzini C, Benedetti L, Civello D, Zanoni C, Rossetti V, Marchesi D, Garavaglia ML, Paulmichl M, Francolini M, Meyer G, Rodighiero S. ICln: a new regulator of non-erythroid 4.1R localisation and function. PLoS One 2014; 9:e108826. [PMID: 25295618 PMCID: PMC4189953 DOI: 10.1371/journal.pone.0108826] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 08/27/2014] [Indexed: 01/12/2023] Open
Abstract
To optimise the efficiency of cell machinery, cells can use the same protein (often called a hub protein) to participate in different cell functions by simply changing its target molecules. There are large data sets describing protein-protein interactions (“interactome”) but they frequently fail to consider the functional significance of the interactions themselves. We studied the interaction between two potential hub proteins, ICln and 4.1R (in the form of its two splicing variants 4.1R80 and 4.1R135), which are involved in such crucial cell functions as proliferation, RNA processing, cytoskeleton organisation and volume regulation. The sub-cellular localisation and role of native and chimeric 4.1R over-expressed proteins in human embryonic kidney (HEK) 293 cells were examined. ICln interacts with both 4.1R80 and 4.1R135 and its over-expression displaces 4.1R from the membrane regions, thus affecting 4.1R interaction with ß-actin. It was found that 4.1R80 and 4.1R135 are differently involved in regulating the swelling activated anion current (ICl,swell) upon hypotonic shock, a condition under which both isoforms are dislocated from the membrane region and thus contribute to ICl,swell current regulation. Both 4.1R isoforms are also differently involved in regulating cell morphology, and ICln counteracts their effects. The findings of this study confirm that 4.1R plays a role in cell volume regulation and cell morphology and indicate that ICln is a new negative regulator of 4.1R functions.
Collapse
Affiliation(s)
- Claudia Bazzini
- Department of Biosciences, University of Milan, Milan, Italy
| | - Lorena Benedetti
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
- Fondazione Filarete for Biosciences and Innovation, Milan, Italy
| | - Davide Civello
- Department of Biosciences, University of Milan, Milan, Italy
| | - Chiara Zanoni
- Pharmaceutical Sciences Department (DISFARM), University of Milan, Milan, Italy
| | | | - Davide Marchesi
- Fondazione Filarete for Biosciences and Innovation, Milan, Italy
| | | | - Markus Paulmichl
- Institute of Pharmacology and Toxicology, Paracelsus Medical University, Salzburg, Austria
| | - Maura Francolini
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
- Fondazione Filarete for Biosciences and Innovation, Milan, Italy
| | - Giuliano Meyer
- Department of Biosciences, University of Milan, Milan, Italy
| | - Simona Rodighiero
- Fondazione Filarete for Biosciences and Innovation, Milan, Italy
- * E-mail:
| |
Collapse
|
6
|
Baines AJ, Lu HC, Bennett PM. The Protein 4.1 family: hub proteins in animals for organizing membrane proteins. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1838:605-19. [PMID: 23747363 DOI: 10.1016/j.bbamem.2013.05.030] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 05/22/2013] [Accepted: 05/28/2013] [Indexed: 01/10/2023]
Abstract
Proteins of the 4.1 family are characteristic of eumetazoan organisms. Invertebrates contain single 4.1 genes and the Drosophila model suggests that 4.1 is essential for animal life. Vertebrates have four paralogues, known as 4.1R, 4.1N, 4.1G and 4.1B, which are additionally duplicated in the ray-finned fish. Protein 4.1R was the first to be discovered: it is a major mammalian erythrocyte cytoskeletal protein, essential to the mechanochemical properties of red cell membranes because it promotes the interaction between spectrin and actin in the membrane cytoskeleton. 4.1R also binds certain phospholipids and is required for the stable cell surface accumulation of a number of erythrocyte transmembrane proteins that span multiple functional classes; these include cell adhesion molecules, transporters and a chemokine receptor. The vertebrate 4.1 proteins are expressed in most tissues, and they are required for the correct cell surface accumulation of a very wide variety of membrane proteins including G-Protein coupled receptors, voltage-gated and ligand-gated channels, as well as the classes identified in erythrocytes. Indeed, such large numbers of protein interactions have been mapped for mammalian 4.1 proteins, most especially 4.1R, that it appears that they can act as hubs for membrane protein organization. The range of critical interactions of 4.1 proteins is reflected in disease relationships that include hereditary anaemias, tumour suppression, control of heartbeat and nervous system function. The 4.1 proteins are defined by their domain structure: apart from the spectrin/actin-binding domain they have FERM and FERM-adjacent domains and a unique C-terminal domain. Both the FERM and C-terminal domains can bind transmembrane proteins, thus they have the potential to be cross-linkers for membrane proteins. The activity of the FERM domain is subject to multiple modes of regulation via binding of regulatory ligands, phosphorylation of the FERM associated domain and differential mRNA splicing. Finally, the spectrum of interactions of the 4.1 proteins overlaps with that of another membrane-cytoskeleton linker, ankyrin. Both ankyrin and 4.1 link to the actin cytoskeleton via spectrin, and we hypothesize that differential regulation of 4.1 proteins and ankyrins allows highly selective control of cell surface protein accumulation and, hence, function. This article is part of a Special Issue entitled: Reciprocal influences between cell cytoskeleton and membrane channels, receptors and transporters. Guest Editor: Jean Claude Hervé
Collapse
Affiliation(s)
| | - Hui-Chun Lu
- Randall Division of Cell and Molecular Biophysics, King's College London, UK
| | - Pauline M Bennett
- Randall Division of Cell and Molecular Biophysics, King's College London, UK.
| |
Collapse
|
7
|
Liu C, Weng H, Chen L, Yang S, Wang H, Debnath G, Guo X, Wu L, Mohandas N, An X. Impaired intestinal calcium absorption in protein 4.1R-deficient mice due to altered expression of plasma membrane calcium ATPase 1b (PMCA1b). J Biol Chem 2013; 288:11407-15. [PMID: 23460639 DOI: 10.1074/jbc.m112.436659] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Protein 4.1R was first identified in the erythrocyte membrane skeleton. It is now known that the protein is expressed in a variety of epithelial cell lines and in the epithelia of many tissues, including the small intestine. However, the physiological function of 4.1R in the epithelial cells of the small intestine has not so far been explored. Here, we show that 4.1R knock-out mice exhibited a significantly impaired small intestinal calcium absorption that resulted in secondary hyperparathyroidism as evidenced by increased serum 1,25-(OH)2-vitamin D3 and parathyroid hormone levels, decreased serum calcium levels, hyperplasia of the parathyroid, and demineralization of the bones. 4.1R is located on the basolateral membrane of enterocytes, where it co-localizes with PMCA1b (plasma membrane calcium ATPase 1b). Expression of PMCA1b in enterocytes was decreased in 4.1(-/-) mice. 4.1R directly associated with PMCA1b, and the association involved the membrane-binding domain of 4.1R and the second intracellular loop and C terminus of PMCA1b. Our findings have enabled us to define a functional role for 4.1R in small intestinal calcium absorption through regulation of membrane expression of PMCA1b.
Collapse
Affiliation(s)
- Congrong Liu
- Red Cell Physiology Laboratory, New York Blood Center, New York, New York 10065, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Pinder JC, Taylor-Harris PM, Bennett PM, Carter E, Hayes NVL, King MDA, Holt MR, Maggs AM, Gascard P, Baines AJ. Isoforms of protein 4.1 are differentially distributed in heart muscle cells: relation of 4.1R and 4.1G to components of the Ca2+ homeostasis system. Exp Cell Res 2012; 318:1467-79. [PMID: 22429617 DOI: 10.1016/j.yexcr.2012.03.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 03/01/2012] [Accepted: 03/02/2012] [Indexed: 11/30/2022]
Abstract
The 4.1 proteins are cytoskeletal adaptor proteins that are linked to the control of mechanical stability of certain membranes and to the cellular accumulation and cell surface display of diverse transmembrane proteins. One of the four mammalian 4.1 proteins, 4.1R (80 kDa/120 kDa isoforms), has recently been shown to be required for the normal operation of several ion transporters in the heart (Stagg MA et al. Circ Res, 2008; 103: 855-863). The other three (4.1G, 4.1N and 4.1B) are largely uncharacterised in the heart. Here, we use specific antibodies to characterise their expression, distribution and novel activities in the left ventricle. We detected 4.1R, 4.1G and 4.1N by immunofluorescence and immunoblotting, but not 4.1B. Only one splice variant of 4.1N and 4.1G was seen whereas there are several forms of 4.1R. 4.1N, like 4.1R, was present in intercalated discs, but unlike 4.1R, it was not localised at the lateral plasma membrane. Both 4.1R and 4.1N were in internal structures that, at the level of resolution of the light microscope, were close to the Z-disc (possibly T-tubules). 4.1G was also in intracellular structures, some of which were coincident with sarcoplasmic reticulum. 4.1G existed in an immunoprecipitable complex with spectrin and SERCA2. 80 kDa 4.1R was present in subcellular fractions enriched in intercalated discs, in a complex resistant to solubilization under non-denaturing conditions. At the intercalated disc 4.1R does not colocalise with the adherens junction protein, β-catenin, but does overlap with the other plasma membrane signalling proteins, the Na/K-ATPase and the Na/Ca exchanger NCX1. We conclude that isoforms of 4.1 proteins are differentially compartmentalised in the heart, and that they form specific complexes with proteins central to cardiomyocyte Ca(2+) metabolism.
Collapse
Affiliation(s)
- Jennifer C Pinder
- King's College London, Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Baklouti F, Morinière M, Haj-Khélil A, Fénéant-Thibault M, Gruffat H, Couté Y, Ninot A, Guitton C, Delaunay J. Homozygous deletion of EPB41 genuine AUG-containing exons results in mRNA splicing defects, NMD activation and protein 4.1R complete deficiency in hereditary elliptocytosis. Blood Cells Mol Dis 2011; 47:158-65. [DOI: 10.1016/j.bcmd.2011.07.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Revised: 07/05/2011] [Indexed: 12/23/2022]
|
10
|
Insights into the Function of the Unstructured N-Terminal Domain of Proteins 4.1R and 4.1G in Erythropoiesis. Int J Cell Biol 2011; 2011:943272. [PMID: 21904552 PMCID: PMC3166722 DOI: 10.1155/2011/943272] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Accepted: 06/20/2011] [Indexed: 01/26/2023] Open
Abstract
Membrane skeletal protein 4.1R is the prototypical member of a family of four highly paralogous proteins that include 4.1G, 4.1N, and 4.1B. Two isoforms of 4.1R (4.1R(135) and 4.1R(80)), as well as 4.1G, are expressed in erythroblasts during terminal differentiation, but only 4.1R(80) is present in mature erythrocytes. One goal in the field is to better understand the complex regulation of cell type and isoform-specific expression of 4.1 proteins. To start answering these questions, we are studying in depth the important functions of 4.1 proteins in the organization and function of the membrane skeleton in erythrocytes. We have previously reported that the binding profiles of 4.1R(80) and 4.1R(135) to membrane proteins and calmodulin are very different despite the similar structure of the membrane-binding domain of 4.1G and 4.1R(135). We have accumulated evidence for those differences being caused by the N-terminal 209 amino acids headpiece region (HP). Interestingly, the HP region is an unstructured domain. Here we present an overview of the differences and similarities between 4.1 isoforms and paralogs. We also discuss the biological significance of unstructured domains.
Collapse
|
11
|
Meyer AJ, Almendrala DK, Go MM, Krauss SW. Structural protein 4.1R is integrally involved in nuclear envelope protein localization, centrosome-nucleus association and transcriptional signaling. J Cell Sci 2011; 124:1433-44. [PMID: 21486941 DOI: 10.1242/jcs.077883] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The multifunctional structural protein 4.1R is required for assembly and maintenance of functional nuclei but its nuclear roles are unidentified. 4.1R localizes within nuclei, at the nuclear envelope, and in cytoplasm. Here we show that 4.1R, the nuclear envelope protein emerin and the intermediate filament protein lamin A/C co-immunoprecipitate, and that 4.1R-specific depletion in human cells by RNA interference produces nuclear dysmorphology and selective mislocalization of proteins from several nuclear subcompartments. Such 4.1R-deficiency causes emerin to partially redistribute into the cytoplasm, whereas lamin A/C is disorganized at nuclear rims and displaced from nucleoplasmic foci. The nuclear envelope protein MAN1, nuclear pore proteins Tpr and Nup62, and nucleoplasmic proteins NuMA and LAP2α also have aberrant distributions, but lamin B and LAP2β have normal localizations. 4.1R-deficient mouse embryonic fibroblasts show a similar phenotype. We determined the functional effects of 4.1R-deficiency that reflect disruption of the association of 4.1R with emerin and A-type lamin: increased nucleus-centrosome distances, increased β-catenin signaling, and relocalization of β-catenin from the plasma membrane to the nucleus. Furthermore, emerin- and lamin-A/C-null cells have decreased nuclear 4.1R. Our data provide evidence that 4.1R has important functional interactions with emerin and A-type lamin that impact upon nuclear architecture, centrosome-nuclear envelope association and the regulation of β-catenin transcriptional co-activator activity that is dependent on β-catenin nuclear export.
Collapse
Affiliation(s)
- Adam J Meyer
- Department of Genome Dynamics, University of California, Berkeley, CA 94720, USA
| | | | | | | |
Collapse
|
12
|
Murata K, Nunomura W, Takakuwa Y, Cherr GN. Two different unique cardiac isoforms of protein 4.1R in zebrafish, Danio rerio, and insights into their cardiac functions as related to their unique structures. Dev Growth Differ 2011; 52:591-602. [PMID: 20887560 DOI: 10.1111/j.1440-169x.2010.01195.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Protein 4.1R (4.1R) has been identified as the major component of the human erythrocyte membrane skeleton. The members of the protein 4.1 gene family are expressed in a tissue-specific alternative splicing manner that increases their functions in each tissue; however, the exact roles of cardiac 4.1R in the developing myocardium are poorly understood. In zebrafish (ZF), we identified two heart-specific 4.1R isoforms, ZF4.1RH2 and ZF4.1RH3, encoding N-terminal 30 kDa (FERM) domain and spectrin-actin binding domain (SABD) and C-terminal domain (CTD), separately. Applying immunohistochemistry using specific antibodies for 30 kDa domain and CTD separately, the gene product of ZF4.1RH2 and ZF4.1RH3 appeared only in the ventricle and in the atrium, respectively, in mature hearts. During embryogenesis, both gene expressions are expressed starting 24 h post-fertilization (hpf). Following whole-mount in situ hybridization, ZF4.1RH3 gene expression was detected in the atrium of 37 hpf embryos. These results indicate that the gene product of ZF4.1RH3 is essential for normal morphological shape of the developing heart and to support the repetitive cycles of its muscle contraction and relaxation.
Collapse
Affiliation(s)
- Kenji Murata
- Department of Animal Science, University of California, Davis, CA, USA.
| | | | | | | |
Collapse
|
13
|
Similarities and differences in the structure and function of 4.1G and 4.1R135, two protein 4.1 paralogues expressed in erythroid cells. Biochem J 2010; 432:407-16. [PMID: 20812914 DOI: 10.1042/bj20100041] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Membrane skeletal protein 4.1R is the prototypical member of a family of four highly paralogous proteins that include 4.1G, 4.1N and 4.1B. Two isoforms of 4.1R (4.1R135 and 4.1R80), as well as 4.1G, are expressed in erythroblasts during terminal differentiation, but only 4.1R80 is present in mature erythrocytes. Although the function of 4.1R isoforms in erythroid cells has been well characterized, there is little or no information on the function of 4.1G in these cells. In the present study, we performed detailed characterization of the interaction of 4.1G with various erythroid membrane proteins and the regulation of these interactions by calcium-saturated calmodulin. Like both isoforms of 4.1R, 4.1G bound to band 3, glycophorin C, CD44, p55 and calmodulin. While both 4.1G and 4.1R135 interact with similar affinity with CD44 and p55, there are significant differences in the affinity of their interaction with band 3 and glycophorin C. This difference in affinity is related to the non-conserved N-terminal headpiece region of the two proteins that is upstream of the 30 kDa membrane-binding domain that harbours the binding sites for the various membrane proteins. The headpiece region of 4.1G also contains a high-affinity calcium-dependent calmodulin-binding site that plays a key role in modulating its interaction with various membrane proteins. We suggest that expression of the two paralogues of protein 4.1 with different affinities for band 3 and glycophorin C is likely to play a role in assembly of these two membrane proteins during terminal erythroid differentiation.
Collapse
|
14
|
Comprehensive characterization of expression patterns of protein 4.1 family members in mouse adrenal gland: implications for functions. Histochem Cell Biol 2010; 134:411-20. [PMID: 20890708 DOI: 10.1007/s00418-010-0749-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2010] [Indexed: 01/22/2023]
Abstract
The members of the protein 4.1 family, 4.1R, 4.1G, 4.1N, and 4.1B, are encoded by four genes, all of which undergo complex alternative splicing. It is well established that 4.1R, the prototypical member of the family, serves as an adapter that links the spectrin-actin based cytoskeleton to the plasma membrane in red cells. It is required for mechanical resilience of the membrane, and it ensures the cell surface accumulation of selected membrane proteins. However, the function of 4.1 proteins outside erythrocytes remains under-explored, especially in endocrine tissues. Transcripts of all 4.1 homologs have previously been documented to be abundantly expressed in adrenal gland. In order to begin to decipher the function of 4.1 proteins in adrenal gland, we performed a detailed characterization of the expression pattern of various 4.1 proteins and their cellular localization. We show that 4.1R (~80 and ~135 kDa) splice forms are expressed on the membrane of all cells, while a ~160 kDa 4.1G splice form is distributed in the cytoplasm and the membrane of zona glomerulosa and of medullary cells. Two 4.1N splice forms, ~135 and ~95 kDa, are present in the peri-nuclear region of both zona glomerulosa and medullary cells, while a single ~130 kDa 4.1B splice form, is detected in all layers of adrenal gland in both the cytoplasm and the membrane. The characterization of distinct splice forms of various 4.1 proteins with diverse cellular and sub-cellular localization indicates multiple functions for this family of proteins in endocrine functions of adrenal gland.
Collapse
|
15
|
Mattagajasingh SN, Huang SC, Benz EJ. Inhibition of protein 4.1 R and NuMA interaction by mutagenization of their binding-sites abrogates nuclear localization of 4.1 R. Clin Transl Sci 2010; 2:102-11. [PMID: 20443879 DOI: 10.1111/j.1752-8062.2008.00087.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Protein 4.1R(4.1R) is a multifunctional structural protein recently implicated in nuclear assembly and cell division. We earlier demonstrated that 4.1R forms a multiprotein complex with mitotic spindle and spindle pole organizing proteins, such as NuMA, dynein, and dynactin, by binding to residues 1788-1810 of NuMA through amino acids encoded by exons 20 and 21 in 24 kD domain. Employing random-and site-directed mutagenesis combined with glycine- and alanine-scanning, we have identified amino acids of 4.1 R and NuMA that sustain their interaction, and have analyzed the effect of mutating the binding sites on their intracellular colocalization. We found that V762, V765, and V767 of 4.1 R, and 11800, 11801,11803, Tl 804, and M1805 of NuMA are necessary for their interaction. GST-fusion peptides of the 4.1R24 kD domain bound to residues 1785-2115 of NuMA in in vitro binding assays, but the binding was inhibited by alanine substitutions of V762, V765, and V767 of 4.1 R, or residues 1800-1805 of NuMA. Additionally, expression of variants of 4.1 R or NuMA that inhibit their in vitro binding also abrogated nuclear localization of 4.1 Rand colocalization with NuMA. Our findings suggest a crucial role of 4.1 R/NuMA interaction in localization and function of 4.1 R in the nucleus.
Collapse
|
16
|
Novel isoform of the Xenopus tropicalis PKA catalytic alpha subunit: An example of alternative splicing. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2010; 5:151-6. [PMID: 20418193 DOI: 10.1016/j.cbd.2010.03.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2010] [Revised: 03/24/2010] [Accepted: 03/27/2010] [Indexed: 11/27/2022]
Abstract
The cAMP-dependent protein kinase (PKA) plays key roles in the control of various aspects of eukaryotic cellular activities by phosphorylating several proteins and is multifunctional in nature. In the case of frog, Xenopus tropicalis, a gene encoding the PKA catalytic alpha subunit has been identified which encodes a single protein. Here we report the occurrence of N-terminal alternative splicing events in X. tropicalis tadpole that, in addition to generating a myristoylatable isoforms, also generate the non-myristoylated variant of the catalytic alpha subunit as has been reported in various other organisms. In addition to the already characterized exon 1, the 5' untranslated region and first intron actually contains one more other exon, that is alternatively spliced on to exon 2 at the 5' end of the pre-mRNA. This N-terminal alternative splicing occurs in combination with already characterized all internal exons. Thus, X. tropicalis tadpole expresses at least two different isoforms of the catalytic alpha subunit of PKA. The significance of this structural diversity in the family of PKA catalytic subunits is discussed.
Collapse
|
17
|
Chromatin architecture and transcription factor binding regulate expression of erythrocyte membrane protein genes. Mol Cell Biol 2009; 29:5399-412. [PMID: 19687298 DOI: 10.1128/mcb.00777-09] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Erythrocyte membrane protein genes serve as excellent models of complex gene locus structure and function, but their study has been complicated by both their large size and their complexity. To begin to understand the intricate interplay of transcription, dynamic chromatin architecture, transcription factor binding, and genomic organization in regulation of erythrocyte membrane protein genes, we performed chromatin immunoprecipitation (ChIP) coupled with microarray analysis and ChIP coupled with massively parallel DNA sequencing in both erythroid and nonerythroid cells. Unexpectedly, most regions of GATA-1 and NF-E2 binding were remote from gene promoters and transcriptional start sites, located primarily in introns. Cooccupancy with FOG-1, SCL, and MTA-2 was found at all regions of GATA-1 binding, with cooccupancy of SCL and MTA-2 also found at regions of NF-E2 binding. Cooccupancy of GATA-1 and NF-E2 was found frequently. A common signature of histone H3 trimethylation at lysine 4, GATA-1, NF-E2, FOG-1, SCL, and MTA-2 binding and consensus GATA-1-E-box binding motifs located 34 to 90 bp away from NF-E2 binding motifs was found frequently in erythroid cell-expressed genes. These results provide insights into our understanding of membrane protein gene regulation in erythropoiesis and the regulation of complex genetic loci in erythroid and nonerythroid cells and identify numerous candidate regions for mutations associated with membrane-linked hemolytic anemia.
Collapse
|
18
|
Baines AJ, Bennett PM, Carter EW, Terracciano C. Protein 4.1 and the control of ion channels. Blood Cells Mol Dis 2009; 42:211-5. [PMID: 19272819 DOI: 10.1016/j.bcmd.2009.01.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2009] [Accepted: 01/20/2009] [Indexed: 11/24/2022]
Abstract
The classical function of 4.1R in red blood cells is to contribute to the mechanochemical properties of the membrane by promoting the interaction between spectrin and actin. More recently, it has been recognized that 4.1R is required for the stable cell surface accumulation of a number of erythrocyte membrane proteins. 4.1R is one member of the mammalian 4.1 family - the others being 4.1N, 4.1G and 4.1B - and is expressed in many cell types other than erythrocytes. Recently we have examined the phenotype of hearts from 4.1R knockout mice. Although they had a generally normal morphology, these hearts exhibited bradycardia, and prolongation of both action potentials and QT intervals. Electrophysiological analysis revealed anomalies in a range of ion channel activities. In addition, the immunoreactivity of voltage-gated Na(+) channel NaV1.5 was reduced, indicating a role for 4.1R in the cellular accumulation of this ion channel. 4.1 proteins also have roles in the accumulation of at least two other classes of ion channel. In epithelia, 4.1 interacts with the store-operated channel TRPC4. In neurons, the ligand-gated channels GluR1 and GluR4 require 4.1 proteins for cell surface accumulation. The spectrum of transmembrane proteins that bind to 4.1 proteins overlaps with that of ankyrin. A hypothesis to investigate in the future is that differential regulation of 4.1 and ankyrins (e.g. by PIP(2)) allows highly selective control of cell surface accumulation and transport activity of a specific range of ion channels.
Collapse
Affiliation(s)
- Anthony J Baines
- Department of Biosciences, University of Kent, Canterbury, CT2 7NJ Kent, UK
| | | | | | | |
Collapse
|
19
|
Marked difference in membrane-protein-binding properties of the two isoforms of protein 4.1R expressed at early and late stages of erythroid differentiation. Biochem J 2009; 417:141-8. [PMID: 18691159 DOI: 10.1042/bj20081372] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Two major isoforms of protein 4.1R, a 135 kDa isoform (4.1R(135)) and an 80 kDa isoform (4.1R(80)), are expressed at distinct stages of terminal erythroid differentiation. The 4.1R(135) isoform is exclusively expressed in early erythroblasts and is not present in mature erythrocytes, whereas the 4.1R(80) isoform is expressed at late stages of erythroid differentiation and is the principal component of mature erythrocytes. These two isoforms differ in that the 4.1R(135) isoform includes an additional 209 amino acids designated as the HP (head-piece) at the N-terminus of 4.1R(80). In the present study, we performed detailed characterization of the interactions of the two 4.1R isoforms with various membrane-binding partners and identified several isoform-specific differences. Although both 4.1R(135) and 4.1R(80) bound to cytoplasmic domains of GPC (glycophorin C) and band 3, there is an order of magnitude difference in the binding affinities. Furthermore, although both isoforms bound CaM (calmodulin), the binding of 4.1R(80) was Ca2+-independent, whereas the binding of 4.1R(135) was strongly Ca2+-dependent. The HP of 4.1R(135) mediates this Ca2+-dependent binding. Ca2+-saturated CaM completely inhibited the binding of 4.1R(135) to GPC, whereas it strongly reduced the affinity of its binding to band 3. Interestingly, in spite of the absence of spectrin-binding activity, the 4.1R(135) isoform was able to assemble on to the membrane of early erythroblasts suggesting that its ability to bind to membrane proteins is sufficient for its membrane localization. These findings enable us to offer potential new insights into the differential contribution of 4.1R isoforms to membrane assembly during terminal erythroid differentiation.
Collapse
|
20
|
Mattagajasingh SN, Huang SC, Benz EJ. Inhibition of Protein 4.1 R and NuMA Interaction by Mutagenization of Their Binding-Sites Abrogates Nuclear Localization of 4.1 R. Clin Transl Sci 2009. [DOI: 10.1111/j.1752-8062.2009.00087.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
21
|
Lospitao E, Pérez-Ferreiro CM, Gosálbez A, Alonso MA, Correas I. An internal ribosome entry site element directs the synthesis of the 80 kDa isoforms of protein 4.1R. BMC Biol 2008; 6:51. [PMID: 19055807 PMCID: PMC2614411 DOI: 10.1186/1741-7007-6-51] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2008] [Accepted: 12/04/2008] [Indexed: 11/12/2022] Open
Abstract
Background In red blood cells, protein 4.1 (4.1R) is an 80 kDa protein that stabilizes the spectrin-actin network and anchors it to the plasma membrane through its FERM domain. While the expression pattern of 4.1R in mature red cells is relatively simple, a rather complex array of 4.1R protein isoforms varying in N-terminal extensions, internal sequences and subcellular locations has been identified in nucleated cells. Among these, 135 kDa and 80 kDa isoforms have different N-terminal extensions and are expressed either from AUG1- or AUG2-containing mRNAs, respectively. These two types of mRNAs, varying solely by presence/absence of 17 nucleotides (nt) which contain the AUG1 codon, are produced by alternative splicing of the 4.1R pre-mRNA. It is unknown whether the 699 nt region comprised between AUG1 and AUG2, kept as a 5' untranslated region in AUG2-containing mRNAs, plays a role on 4.1R mRNA translation. Results By analyzing the in vitro expression of a panel of naturally occurring 4.1R cDNAs, we observed that all AUG1/AUG2-containing cDNAs gave rise to both long, 135 kDa, and short, 80 kDa, 4.1R isoforms. More importantly, similar results were also observed in cells transfected with this set of 4.1R cDNAs. Mutational studies indicated that the short isoforms were not proteolytic products of the long isoforms but products synthesized from AUG2. The presence of a cryptic promoter in the 4.1R cDNA sequence was also discounted. When a 583 nt sequence comprised between AUG1 and AUG2 was introduced into bicistronic vectors it directed protein expression from the second cistron. This was also the case when ribosome scanning was abolished by introduction of a stable hairpin at the 5' region of the first cistron. Deletion analysis of the 583 nt sequence indicated that nucleotides 170 to 368 are essential for expression of the second cistron. The polypyrimidine tract-binding protein bound to the 583 nt active sequence but not to an inactive 3'-fragment of 149 nucleotides. Conclusion Our study is the first demonstration of an internal ribosome entry site as a mechanism ensuring the production of 80 kDa isoforms of protein 4.1R. This mechanism might also account for the generation of 60 kDa isoforms of 4.1R from a downstream AUG3. Our results reveal an additional level of control to 4.1R gene expression pathways and will contribute to the understanding of the biology of proteins 4.1R and their homologues, comprising an ample family of proteins involved in cytoskeletal organization.
Collapse
Affiliation(s)
- Eva Lospitao
- Departamento de Biología Molecular, Universidad Autónoma de Madrid y Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Nicolás Cabrera, Madrid, Spain.
| | | | | | | | | |
Collapse
|
22
|
Seo PS, Jeong JJ, Zeng L, Takoudis CG, Quinn BJ, Khan AA, Hanada T, Chishti AH. Alternatively spliced exon 5 of the FERM domain of protein 4.1R encodes a novel binding site for erythrocyte p55 and is critical for membrane targeting in epithelial cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2008; 1793:281-9. [PMID: 18952129 DOI: 10.1016/j.bbamcr.2008.09.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2008] [Revised: 09/12/2008] [Accepted: 09/17/2008] [Indexed: 01/18/2023]
Abstract
Direct physical linkage of MAGUKs to the actin cytoskeleton was first established by the interaction of erythrocyte p55 with the FERM domain of protein 4.1R. Subsequently, it was reported that p55 binds to a 51-amino acid peptide, encoded by exon 10, located within the FERM domain of protein 4.1R. In this study, we investigated the nature of the p55-FERM domain binding interface and show that p55 binds to a second 35-amino acid peptide, encoded by an alternatively spliced exon 5, located within the FERM domain of protein 4.1R. Competition and Surface Plasmon Resonance-binding measurements suggest that the peptides encoded by exons 5 and 10 bind to independent sites within the D5 domain of p55. Interestingly, the full length 135 kDa isoform of protein 4.1R containing both exons 5 and 10 was targeted exclusively to the plasma membrane of epithelial cells whereas the same isoform without exon 5 completely lost its membrane localization capacity. Together, these results indicate that p55 binds to two distinct sites within the FERM domain, and the alternatively spliced exon 5 is necessary for the membrane targeting of protein 4.1R in epithelial cells. Since sequences similar to the exon 5-peptide of protein 4.1R and D5 domain of p55 are conserved in many proteins, our findings suggest that a similar mechanism may govern the membrane targeting of other FERM domain containing proteins.
Collapse
Affiliation(s)
- Pil-Soo Seo
- Department of Pharmacology, UIC Cancer Center, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Alternative spliced variants in the pantetheinase family of genes expressed in human neutrophils. Gene 2008; 426:57-64. [PMID: 18805469 DOI: 10.1016/j.gene.2008.08.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2008] [Revised: 08/06/2008] [Accepted: 08/22/2008] [Indexed: 11/21/2022]
Abstract
Pantetheinase (EC 3.5.1.92) is an enzyme that hydrolyzes pantetheine, an intermediate metabolite of coenzyme A, into pantothenic acid (vitamin B(5)) and cysteamine, a potent antioxidant. The pantetheinase gene family consists of three independent genes, pantetheinase/vanin-1/VNN1, GPI-80/VNN2 and vanin-3/VNN3 that are each composed of seven exons. We herein report that human neutrophils express transcripts encoding at least nine splice variants of VNN3 and four splice variants of GPI-80/VNN2. Analysis of the DNA sequence of the human VNN3 gene demonstrated that the VNN3 locus in the human genome as well as the sequence of cDNA clones obtained in this study does not encode the complete VNN3 protein, as previously reported due to a frame shift caused by lack of one nucleotide. Moreover, the VNN3 locus indeed encodes smaller peptides compared to the proteins encoded by the mouse orthologous gene, vanin-3. The anti-GPI-80 monoclonal antibody 3H9 recognized amino acids 120-179 of the GPI-80/VNN2 protein as shown by the results of immunoblotting with recombinant GPI-80/VNN2 variant proteins. Immunoblotting with human neutrophil lysate suggests that the GPI-80/VNN2 variants exist in human neutrophils. The existence of splice variants in the pantetheinase gene family suggests the possibility of alternative roles in addition to canonical enzymatic activity in human neutrophils.
Collapse
|
24
|
Identification of CTLA-4 isoforms produced by alternative splicing and their association with myasthenia gravis. Clin Immunol 2008; 128:374-81. [DOI: 10.1016/j.clim.2008.05.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2007] [Revised: 04/19/2008] [Accepted: 05/19/2008] [Indexed: 11/23/2022]
|
25
|
Nunomura W, Takakuwa Y, Cherr GN, Murata K. Characterization of protein 4.1R in erythrocytes of zebrafish (Danio rerio): unique binding properties with transmembrane proteins and calmodulin. Comp Biochem Physiol B Biochem Mol Biol 2007; 148:124-38. [PMID: 17569566 DOI: 10.1016/j.cbpb.2007.05.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2006] [Revised: 05/03/2007] [Accepted: 05/05/2007] [Indexed: 11/17/2022]
Abstract
Cytoskeletal protein 4.1R is instrumental in regulating erythrocyte plasticity. 4.1R is comprised of four domains identified after chymotryptic digestion: an N-terminal 30 kDa domain responsible for interaction with membrane proteins, a unique domain, a spectrin-actin binding (SAB) domain, and a C-terminal domain (CTD). 4.1R 30 kDa domain interactions with transmembrane proteins are regulated by the Ca(2+)/calmodulin (CaM) complex. Unlike mature mammalian erythrocytes, fish erythrocytes remain nucleated. Comparing their cytoskeleton architecture and functional properties is therefore of great interest. Here we characterized the recently cloned zebrafish (Danio rerio, ZF) 4.1R and compared its properties with human 4.1R. We identified three ZF4.1R mRNA transcripts in erythrocytes, all characterized by exclusion of the central domains. The major transcript, referred to as BL31, included a full length 30 kDa domain (ZFR30) and parts of the unique region Ua and of CTD. Two minor transcripts, referred to as BL42 and BL56, expressed parts of ZFR30 and of the unique region Ub and full length SAB and CTD domains. Antibodies to ZFR30, ZF4.1R CTD and ZF glycophorin C (GPC) labeled the ZF erythrocyte plasma membrane. ZFR30 bound to CaM in presence or absence of Ca(2+). Resonant mirror detection binding assays revealed that ZFR30 bound to human Band3 with low K((D)) ( approximately 10nM), and to GPC with higher K((D)) ( approximately 1nM). The Ca(2+)/CaM complex did not affect ZFR30 binding to Band3 and GPC. Finally, we confirmed ZFR30 binding to erythrocyte plasma membrane proteins by pulling down ZFR30 with human erythrocyte inside-out vesicles (IOV). This study defines unique structural and functional properties for ZF4.1R.
Collapse
Affiliation(s)
- Wataru Nunomura
- Department of Biochemistry, Tokyo Women's Medical University, Kawada 8-1, Shinjuku, Tokyo 162-8666, Japan.
| | | | | | | |
Collapse
|
26
|
Pérez-Ferreiro CM, Lospitao E, Correas I. Protein 4.1R self-association: identification of the binding domain. Biochem J 2006; 400:457-65. [PMID: 16881872 PMCID: PMC1698608 DOI: 10.1042/bj20060644] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Erythroid protein 4.1 (4.1R) stabilizes the spectrin-actin network and anchors it to the plasma membrane. To contribute to the characterization of non-erythroid protein 4.1R, we used sedimentation, pull-down and co-immunoprecipitation assays to investigate the ability of protein 4.1R to establish inter-/intra-molecular associations. We demonstrated that the small 4.1R isoforms of 60 kDa (4.1R60), but not the larger isoforms of 80 and 135 kDa (4.1R80 and 4.1R135), were self-associated, and that a domain contained in all 4.1R isoforms, the core region, was responsible for 4.1R self-association. Results from denaturing-renaturing experiments, in which an initially non-self-associated 4.1R80 isoform became self-associated, suggested that an initially hidden core region was subsequently exposed. This hypothesis was supported by results from pull-down assays, which showed that the core region interacted with the N-terminal end of the FERM (4.1, ezrin, radixin, moesin) domain that is present in 4.1R80 and 4.1R135 isoforms but absent from 4.1R60 isoforms. Consistently, 4.1R80 isoforms bound neither to each other nor to 4.1R60 isoforms. We propose that 4.1R60 isoforms are constitutively self-associated, whereas 4.1R80 and 4.1R135 self-association is prevented by intramolecular interactions.
Collapse
Affiliation(s)
- Carmen M Pérez-Ferreiro
- Departamento de Biología Molecular, CBMSO (Centro de Biología Molecular Severo Ochoa), UAM/CSIC, Cantoblanco, E-28049 Madrid, Spain.
| | | | | |
Collapse
|
27
|
García-Sacristán A, Fernández-Nestosa MJ, Hernández P, Schvartzman JB, Krimer DB. Protein kinase clk/STY is differentially regulated during erythroleukemia cell differentiation: a bias toward the skipped splice variant characterizes postcommitment stages. Cell Res 2005; 15:495-503. [PMID: 16045812 DOI: 10.1038/sj.cr.7290319] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Clk/STY is a LAMMER protein kinase capable to phosphorylate serine/arginine-rich (SR) proteins that modulate pre-mRNA splicing. Clk/STY alternative splicing generates transcripts encoding a full-length kinase and a truncated catalytically inactive protein. Here we showed that clk/STY, as well as other members of the family (e.g. clk2, clk3 and clk4), are up-regulated during HMBA-induced erythroleukemia cell differentiation. mRNAs coding for the full-length and the truncated forms were responsible for the overall increased expression. In clk/STY, however, a switch was observed for the ratio of the two alternative spliced products. In undifferentiated cells the full-length transcript was more abundant whereas the transcript encoding for the truncated form predominated at latter stages of differentiation. Surprisingly, overexpression of clk/STY did not alter the splicing switch upon differentiation in MEL cells. These results suggest that clk/STY might contribute to control erythroid differentiation by a mechanism that implicates a balance between these two isoforms.
Collapse
Affiliation(s)
- Ana García-Sacristán
- Department of Cell and Developmental Biology, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, Madrid 28040, Spain
| | | | | | | | | |
Collapse
|
28
|
Crable SC, Hammond SM, Papes R, Rettig RK, Zhou GP, Gallagher PG, Joiner CH, Anderson KP. Multiple isoforms of the KC1 cotransporter are expressed in sickle and normal erythroid cells. Exp Hematol 2005; 33:624-31. [PMID: 15911086 DOI: 10.1016/j.exphem.2005.02.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2005] [Revised: 02/24/2005] [Accepted: 02/28/2005] [Indexed: 11/22/2022]
Abstract
OBJECTIVE The KCl cotransporter (KCC) plays an important role in cellular cation and volume regulation and contributes to the process of volume reduction that accompanies reticulocyte maturation. In human red cells containing sickle hemoglobin, KCl cotransporter activity is high compared to normal cells, and contributes to the deleterious dehydration of sickle reticulocytes. To date, genes for four KCC isoforms have been identified. As a step toward determining which isoform(s) is responsible for the Cl-dependent K fluxes in reticulocytes, human erythroid cells were examined for the presence of various KCC isoform transcripts. METHODS In vitro differentiated erythroid precursors, and reticulocytes isolated from normal individuals and sickle patients, were examined by reverse-transcriptase PCR for the expression of KCC isoforms. Transient transfection experiments were subsequently performed to characterize a novel KCC1 promoter. RESULTS Expression of multiple isoforms was detected, with transcripts for KCC1, 3, and 4 detected in all samples of erythroid cells. Two N-terminal splicing variants were detected for both KCC1 and 3. Sickle hemoglobin containing reticulocytes demonstrated KCC isoform expression patterns similar to wild-type cells, except for a consistent difference in the relative abundance of one KCC1 splice variant. This N-terminal variant initiates from a newly described promoter in the KCC1 gene. CONCLUSION Three KCC genes are expressed in human red cells. Splicing variants arising from the KCC1 and 3 genes are also evident. Structure/function studies of mouse KCC1 suggest that these natural variants could profoundly affect overall cotransporter activity in the red cell.
Collapse
Affiliation(s)
- Scott C Crable
- Comprehensive Sickle Cell Center, Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio 45229, USA
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Gascard P, Parra MK, Zhao Z, Calinisan VR, Nunomura W, Rivkees SA, Mohandas N, Conboy JG. Putative tumor suppressor protein 4.1B is differentially expressed in kidney and brain via alternative promoters and 5' alternative splicing. ACTA ACUST UNITED AC 2004; 1680:71-82. [PMID: 15488987 DOI: 10.1016/j.bbaexp.2004.08.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2004] [Revised: 08/03/2004] [Accepted: 08/10/2004] [Indexed: 10/26/2022]
Abstract
Protein 4.1B has been reported as a tumor suppressor in brain, but not in kidney, despite high expression in both tissues. Here we demonstrate that N-terminal variability in kidney and brain 4.1B isoforms arises through an unusual coupling of RNA processing events in the 5' region of the gene. We describe two transcriptional promoters at far upstream alternative exons 1A and 1B, and show that their respective transcripts splice differentially to exon 2'/2 in a manner that determines mRNA coding capacity. The consequence of this unique processing is that exon 1B transcripts initiate translation at AUG1 (in exon 2') and encode larger 4.1B isoforms with an N-terminal extension; exon 1A transcripts initiate translation at AUG2 (in exon 4) and encode smaller 4.1B isoforms. Tissue-specific differences in promoter utilization may thus explain the abundance of larger 4.1B isoforms in brain but not in kidney. In cell studies, differentiation of PC12 cells was accompanied by translocation of large protein 4.1B isoforms into the nucleus. We propose that first exon specification is coupled to downstream splicing events, generating 4.1B isoforms with diverse roles in kidney and brain physiology, and potentially unique functions in cell proliferation and tumor suppression.
Collapse
Affiliation(s)
- Philippe Gascard
- Life Sciences Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Mail stop 74-157, Berkeley CA 94720, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Huang SC, Liu ES, Chan SH, Munagala ID, Cho HT, Jagadeeswaran R, Benz EJ. Mitotic regulation of protein 4.1R involves phosphorylation by cdc2 kinase. Mol Biol Cell 2004; 16:117-27. [PMID: 15525677 PMCID: PMC539157 DOI: 10.1091/mbc.e04-05-0426] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The nonerythrocyte isoform of the cytoskeletal protein 4.1R (4.1R) is associated with morphologically dynamic structures during cell division and has been implicated in mitotic spindle function. In this study, we define important 4.1R isoforms expressed in interphase and mitotic cells by RT-PCR and mini-cDNA library construction. Moreover, we show that 4.1R is phosphorylated by p34cdc2 kinase on residues Thr60 and Ser679 in a mitosis-specific manner. Phosphorylated 4.1R135 isoform(s) associate with tubulin and Nuclear Mitotic Apparatus protein (NuMA) in intact HeLa cells in vivo as well as with the microtubule-associated proteins in mitotic asters assembled in vitro. Recombinant 4.1R135 is readily phosphorylated in mitotic extracts and reconstitutes mitotic aster assemblies in 4.1R-immunodepleted extracts in vitro. Furthermore, phosphorylation of these residues appears to be essential for the targeting of 4.1R to the spindle poles and for mitotic microtubule aster assembly in vitro. Phosphorylation of 4.1R also enhances its association with NuMA and tubulin. Finally, we used siRNA inhibition to deplete 4.1R from HeLa cells and provide the first direct genetic evidence that 4.1R is required to efficiently focus mitotic spindle poles. Thus, we suggest that 4.1R is a member of the suite of direct cdc2 substrates that are required for the establishment of a bipolar spindle.
Collapse
Affiliation(s)
- Shu-Ching Huang
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA 02115, USA.
| | | | | | | | | | | | | |
Collapse
|
31
|
Parra M, Gee S, Chan N, Ryaboy D, Dubchak I, Mohandas N, Gascard PD, Conboy JG. Differential domain evolution and complex RNA processing in a family of paralogous EPB41 (protein 4.1) genes facilitate expression of diverse tissue-specific isoforms. Genomics 2004; 84:637-46. [PMID: 15475241 DOI: 10.1016/j.ygeno.2004.06.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2004] [Accepted: 06/08/2004] [Indexed: 11/30/2022]
Abstract
The EPB41 (protein 4.1) genes epitomize the resourcefulness of the mammalian genome to encode a complex proteome from a small number of genes. By utilizing alternative transcriptional promoters and tissue-specific alternative pre-mRNA splicing, EPB41, EPB41L2, EPB41L3, and EPB41L1 encode a diverse array of structural adapter proteins. Comparative genomic and transcript analysis of these 140- to 240-kb genes indicates several unusual features: differential evolution of highly conserved exons encoding known functional domains interspersed with unique exons whose size and sequence variations contribute substantially to intergenic diversity; alternative first exons, most of which map far upstream of the coding regions; and complex tissue-specific alternative pre-mRNA splicing that facilitates synthesis of functionally different complements of 4.1 proteins in various cells. Understanding the splicing regulatory networks that control protein 4.1 expression will be critical to a full appreciation of the many roles of 4.1 proteins in normal cell biology and their proposed roles in human cancer.
Collapse
Affiliation(s)
- Marilyn Parra
- Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Krauss SW, Lee G, Chasis JA, Mohandas N, Heald R. Two Protein 4.1 Domains Essential for Mitotic Spindle and Aster Microtubule Dynamics and Organization in Vitro. J Biol Chem 2004; 279:27591-8. [PMID: 15102852 DOI: 10.1074/jbc.m402813200] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Multifunctional structural proteins belonging to the 4.1 family are components of nuclei, spindles, and centrosomes in vertebrate cells. Here we report that 4.1 is critical for spindle assembly and the formation of centrosome-nucleated and motor-dependent self-organized microtubule asters in metaphase-arrested Xenopus egg extracts. Immunodepletion of 4.1 disrupted microtubule arrays and mislocalized the spindle pole protein NuMA. Remarkably, assembly was completely rescued by supplementation with a recombinant 4.1R isoform. We identified two 4.1 domains critical for its function in microtubule polymerization and organization utilizing dominant negative peptides. The 4.1 spectrin-actin binding domain or NuMA binding C-terminal domain peptides caused morphologically disorganized structures. Control peptides with low homology or variant spectrin-actin binding domain peptides that were incapable of binding actin had no deleterious effects. Unexpectedly, the addition of C-terminal domain peptides with reduced NuMA binding caused severe microtubule destabilization in extracts, dramatically inhibiting aster and spindle assembly and also depolymerizing preformed structures. However, the mutant C-terminal peptides did not directly inhibit or destabilize microtubule polymerization from pure tubulin in a microtubule pelleting assay. Our data showing that 4.1 is a crucial factor for assembly and maintenance of mitotic spindles and self-organized and centrosome-nucleated microtubule asters indicates that 4.1 is involved in regulating both microtubule dynamics and organization. These investigations underscore an important functional context for protein 4.1 in microtubule morphogenesis and highlight a previously unappreciated role for 4.1 in cell division.
Collapse
Affiliation(s)
- Sharon Wald Krauss
- Lawrence Berkeley National Laboratory, Life Sciences Division, University of California, Berkeley 94720, USA.
| | | | | | | | | |
Collapse
|
33
|
Abstract
Hereditary elliptocytosis (HE) is a common disorder of erythrocyte shape, occurring especially in individuals of African and Mediterranean ancestry, presumably because elliptocytes confer some resistance to malaria. The principle lesion in HE is mechanical weakness or fragility of the erythrocyte membrane skeleton due to defects in alpha-spectrin, beta-spectrin, or protein 4.1. Numerous mutations have been described in the genes encoding these proteins, including point mutations, gene deletions and insertions, and mRNA processing defects. Several mutations have been identified in a number of individuals on the same genetic background, suggesting a "founder effect." The majority of HE patients are asymptomatic, but some may experience hemolytic anemia, splenomegaly, and intermittent jaundice.
Collapse
Affiliation(s)
- Patrick G Gallagher
- Department of Pediatrics, Yale University School of Medicine, 333 Cedar Street, PO Box 208064, New Haven, CT 06520-8064, USA
| |
Collapse
|
34
|
Kittiniyom K, Mastronardi M, Roemer M, Wells WA, Greenberg ER, Titus-Ernstoff L, Newsham IF. Allele-specific loss of heterozygosity at theDAL-1/4.1B (EPB41L3) tumor-suppressor gene locus in the absence of mutation. Genes Chromosomes Cancer 2004; 40:190-203. [PMID: 15138999 DOI: 10.1002/gcc.20034] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
DAL-1/4.1B (EPB41L3)is a member of the protein 4.1 superfamily, which encompasses structural proteins that play important roles in membrane processes via interactions with actin, spectrin, and the cytoplasmic domains of integral membrane proteins. DAL-1/4.1B localizes within chromosomal region 18p11.3, which is affected by loss of heterozygosity (LOH) in various adult tumors. Reintroduction of this protein into DAL-1/4.1B-null lung and breast tumor cell lines significantly reduced the number of cells, providing functional evidence that this protein possesses a growth suppressor function not confined to a single cell type. For characterization of the mutational mechanisms responsible for loss of DAL-1/4.1B function in tumors, the exon-intron structure of DAL-1/4.1B was examined for mutations in 15 normal/tumor pairs of non-small cell lung carcinoma by single-strand conformation polymorphism analysis. These studies revealed that small intragenic mutations are uncommon in DAL-1/4.1B. Furthermore, LOH analysis on 129 informative early-stage breast tumors utilizing a new intragenic C/T single-nucleotide polymorphism in exon 14 revealed that LOH resulted in preferential retention of the C-containing allele, suggesting that allele-specific loss is occurring. These studies indicate that mechanisms such as imprinting or monoallelic expression in combination with loss of heterozygosity may be responsible for loss of the DAL-1/4.1B protein in early breast disease.
Collapse
Affiliation(s)
- Kanokwan Kittiniyom
- David and Doreen Hermelin Laboratory of Molecular Oncogenetics, Hermelin Brain Tumor Center and Department of Neurosurgery, Henry Ford Hospital, Detroit, Michigan 48202, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
Andersen JS, Wilkinson CJ, Mayor T, Mortensen P, Nigg EA, Mann M. Proteomic characterization of the human centrosome by protein correlation profiling. Nature 2003; 426:570-4. [PMID: 14654843 DOI: 10.1038/nature02166] [Citation(s) in RCA: 1026] [Impact Index Per Article: 46.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2003] [Accepted: 10/30/2003] [Indexed: 11/08/2022]
Abstract
The centrosome is the major microtubule-organizing centre of animal cells and through its influence on the cytoskeleton is involved in cell shape, polarity and motility. It also has a crucial function in cell division because it determines the poles of the mitotic spindle that segregates duplicated chromosomes between dividing cells. Despite the importance of this organelle to cell biology and more than 100 years of study, many aspects of its function remain enigmatic and its structure and composition are still largely unknown. We performed a mass-spectrometry-based proteomic analysis of human centrosomes in the interphase of the cell cycle by quantitatively profiling hundreds of proteins across several centrifugation fractions. True centrosomal proteins were revealed by both correlation with already known centrosomal proteins and in vivo localization. We identified and validated 23 novel components and identified 41 likely candidates as well as the vast majority of the known centrosomal proteins in a large background of nonspecific proteins. Protein correlation profiling permits the analysis of any multiprotein complex that can be enriched by fractionation but not purified to homogeneity.
Collapse
Affiliation(s)
- Jens S Andersen
- Center for Experimental BioInformatics, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | | | | | | | | | | |
Collapse
|
36
|
Lee JCM, Gimm JA, Lo AJ, Koury MJ, Krauss SW, Mohandas N, Chasis JA. Mechanism of protein sorting during erythroblast enucleation: role of cytoskeletal connectivity. Blood 2003; 103:1912-9. [PMID: 14563645 DOI: 10.1182/blood-2003-03-0928] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
During erythroblast enucleation, nuclei surrounded by plasma membrane separate from erythroblast cytoplasm. A key aspect of this process is sorting of erythroblast plasma membrane components to reticulocytes and expelled nuclei. Although it is known that cytoskeletal elements actin and spectrin partition to reticulocytes, little is understood about molecular mechanisms governing plasma membrane protein sorting. We chose glycophorin A (GPA) as a model integral protein to begin investigating protein-sorting mechanisms. Using immunofluorescence microscopy and Western blotting we found that GPA sorted predominantly to reticulocytes. We hypothesized that the degree of skeletal linkage might control the sorting pattern of transmembrane proteins. To explore this hypothesis, we quantified the extent of GPA association to the cytoskeleton in erythroblasts, young reticulocytes, and mature erythrocytes using fluorescence imaged microdeformation (FIMD) and observed that GPA underwent dramatic reorganization during terminal differentiation. We discovered that GPA was more connected to the membrane cytoskeleton, either directly or indirectly, in erythroblasts and young reticulocytes than in mature cells. We conclude that skeletal protein association can regulate protein sorting during enucleation. Further, we suggest that the enhanced rigidity of reticulocyte membranes observed in earlier investigations results, at least in part, from increased connectivity of GPA with the spectrin-based skeleton.
Collapse
Affiliation(s)
- James C-M Lee
- Department of Biological Engineering, University of Missouri-Columbia, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Robb VA, Li W, Gascard P, Perry A, Mohandas N, Gutmann DH. Identification of a third Protein 4.1 tumor suppressor, Protein 4.1R, in meningioma pathogenesis. Neurobiol Dis 2003; 13:191-202. [PMID: 12901833 DOI: 10.1016/s0969-9961(03)00071-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Meningiomas are common central nervous system tumors; however, the mechanisms underlying their pathogenesis are largely undefined. In this report, we demonstrate that a third Protein 4.1 family member, Protein 4.1R, functions as a meningioma tumor suppressor. We observed loss of Protein 4.1R expression in two meningioma cell lines (IOMM-Lee, CH157-MN) by Western blotting as well as in 6 of 15 sporadic meningiomas by immunohistochemistry and fluorescence in situ hybridization. In support of a meningioma tumor suppressor function, Protein 4.1R overexpression resulted in reduced IOMM-Lee and CH157-MN cell proliferation. Similar to the Protein 4.1B and merlin tumor suppressors, Protein 4.1R membrane localization increased significantly under conditions of growth arrest in vitro. Lastly, we show that Protein 4.1R interacted with a subset of merlin/Protein 4.1B interactors including CD44 and betaII-spectrin. Collectively, these results suggest that Protein 4.1R functions as an important tumor suppressor in the molecular pathogenesis of meningioma.
Collapse
Affiliation(s)
- Victoria A Robb
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | | |
Collapse
|
38
|
Parra MK, Gee SL, Koury MJ, Mohandas N, Conboy JG. Alternative 5' exons and differential splicing regulate expression of protein 4.1R isoforms with distinct N-termini. Blood 2003; 101:4164-71. [PMID: 12522012 DOI: 10.1182/blood-2002-06-1796] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Among the alternative pre-mRNA splicing events that characterize protein 4.1R gene expression, one involving exon 2' plays a critical role in regulating translation initiation and N-terminal protein structure. Exon 2' encompasses translation initiation site AUG1 and is located between alternative splice acceptor sites at the 5' end of exon 2; its inclusion or exclusion from mature 4.1R mRNA regulates expression of longer or shorter isoforms of 4.1R protein, respectively. The current study reports unexpected complexity in the 5' region of the 4.1R gene that directly affects alternative splicing of exon 2'. Identified far upstream of exon 2 in both mouse and human genomes were 3 mutually exclusive alternative 5' exons, designated 1A, 1B, and 1C; all 3 are associated with strong transcriptional promoters in the flanking genomic sequence. Importantly, exons 1A and 1B splice differentially with respect to exon 2', generating transcripts with different 5' ends and distinct N-terminal protein coding capacity. Exon 1A-type transcripts splice so as to exclude exon 2' and therefore utilize the downstream AUG2 for translation of 80-kDa 4.1R protein, whereas exon 1B transcripts include exon 2' and initiate at AUG1 to synthesize 135-kDa isoforms. RNA blot analyses revealed that 1A transcripts increase in abundance in late erythroblasts, consistent with the previously demonstrated up-regulation of 80-kDa 4.1R during terminal erythroid differentiation. Together, these results suggest that synthesis of structurally distinct 4.1R protein isoforms in various cell types is regulated by a novel mechanism requiring coordination between upstream transcription initiation events and downstream alternative splicing events.
Collapse
Affiliation(s)
- Marilyn K Parra
- Lawrence Berkeley National Laboratory, Life Sciences Division, Berkeley, CA 94720, USA
| | | | | | | | | |
Collapse
|
39
|
Ramez M, Blot-Chabaud M, Cluzeaud F, Chanan S, Patterson M, Walensky LD, Marfatia S, Baines AJ, Chasis JA, Conboy JG, Mohandas N, Gascard P. Distinct distribution of specific members of protein 4.1 gene family in the mouse nephron. Kidney Int 2003; 63:1321-37. [PMID: 12631349 DOI: 10.1046/j.1523-1755.2003.00870.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Protein 4.1 is an adapter protein that links the actin cytoskeleton to various transmembrane proteins. These 4.1 proteins are encoded by four homologous genes, 4.1R, 4.1G, 4.1N, and 4.1B, which undergo complex alternative splicing. Here we performed a detailed characterization of the expression of specific 4.1 proteins in the mouse nephron. METHODS Distribution of renal 4.1 proteins was investigated by staining of paraformaldehyde-fixed mouse kidney sections with antibodies highly specific for each 4.1 protein. Major 4.1 splice forms, amplified from mouse kidney marathon cDNA, were expressed in transfected COS-7 cells in order to assign species of known exon composition to proteins detected in kidney. RESULTS A 105 kD 4.1R splice form, initiating at ATG-2 translation initiation site and lacking exon 16, but including exon 17B, was restricted to thick ascending limb of Henle's loop. A 95 kD 4.1N splice form, lacking exons 15 and 17D, was expressed in either descending or ascending thin limb of Henle's loop, distal convoluted tubule, and all regions of the collecting duct system. A major 108 kD 4.1B splice form, initiating at a newly characterized ATG translation initiation site, and lacking exons 15, 17B, and 21, was present only in Bowman's capsule and proximal convoluted tubule (PCT). There was no expression of 4.1G in kidney. CONCLUSION Distinct distribution of 4.1 proteins along the nephron suggests their involvement in targeting of selected transmembrane proteins in kidney epithelium and, therefore, in regulation of specific kidney functions.
Collapse
Affiliation(s)
- Mohamed Ramez
- Department of Subcellular Structure, Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Proteins of the erythrocyte membrane have served as the prototypes of homologous families of multifunctional proteins in erythroid and nonerythroid cells. These proteins demonstrate many different cell type, tissue-specific, and developmental stage-specific functions. This complex pattern of functional diversity appears to have evolved from the cell type, tissue-specific, developmentally regulated expression of multiple protein isoforms. Isoform diversity arises from different gene products from related genes; from differential, alternate splicing of the same gene product; from the use of tissue-specific promoters; and from alternate polyadenylation. The identification and characterization of the regulatory elements that control erythrocyte membrane protein gene expression have important implications for several biologic processes. These include disease pathogenesis, membrane assembly, hematopoiesis, gene regulation, and direction of other erythroid-specific genes in transgenic mouse and gene therapy applications.
Collapse
Affiliation(s)
- Patrick G Gallagher
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut 06520-8064, USA.
| |
Collapse
|
41
|
Luque CM, Pérez-Ferreiro CM, Pérez-Gonzalez A, Englmeier L, Koffa MD, Correas I. An alternative domain containing a leucine-rich sequence regulates nuclear cytoplasmic localization of protein 4.1R. J Biol Chem 2003; 278:2686-91. [PMID: 12427749 DOI: 10.1074/jbc.m201521200] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
In red blood cells, protein 4.1 (4.1R) is an 80-kDa protein that stabilizes the spectrin-actin network and anchors it to the plasma membrane. The picture is more complex in nucleated cells, in which many 4.1R isoforms, varying in size and intracellular location, have been identified. To contribute to the characterization of signals involved in differential intracellular localization of 4.1R, we have analyzed the role the exon 5-encoded sequence plays in 4.1R distribution. We show that exon 5 encodes a leucine-rich sequence that shares key features with nuclear export signals (NESs). This sequence adopts the topology employed for NESs of other proteins and conserves two hydrophobic residues that are shown to be critical for NES function. A 4.1R isoform expressing the leucine-rich sequence binds to the export receptor CRM1 in a RanGTP-dependent fashion, whereas this does not occur in a mutant whose two conserved hydrophobic residues are substituted. These two residues are also essential for 4.1R intracellular distribution, because the 4.1R protein containing the leucine-rich sequence localizes in the cytoplasm, whereas the mutant protein predominantly accumulates in the nucleus. We hypothesize that the leucine-rich sequence in 4.1R controls distribution and concomitantly function of a specific set of 4.1R isoforms.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Binding Sites
- COS Cells
- Cell Nucleus/metabolism
- Cloning, Molecular
- Cytoplasm/metabolism
- Cytoskeletal Proteins
- DNA, Complementary/metabolism
- Exons
- Green Fluorescent Proteins
- Humans
- Karyopherins/metabolism
- Leucine/chemistry
- Leucine/metabolism
- Luminescent Proteins/metabolism
- Membrane Proteins
- Microscopy, Fluorescence
- Models, Biological
- Models, Genetic
- Models, Molecular
- Molecular Sequence Data
- Mutagenesis, Site-Directed
- Mutation
- Neuropeptides
- Protein Biosynthesis
- Protein Conformation
- Protein Isoforms
- Protein Structure, Secondary
- Protein Structure, Tertiary
- Receptors, Cytoplasmic and Nuclear
- Transfection
- ran GTP-Binding Protein/metabolism
- Exportin 1 Protein
Collapse
Affiliation(s)
- Carlos M Luque
- Departamento de Biologia Molecular, Centro de Biologia Molecular Severo Ochoa (Consejo Superior de Investigaciones Cientificas/Universidad Autónoma de Madrid), Universidad Autónoma de Madrid, Spain
| | | | | | | | | | | |
Collapse
|
42
|
Veistinen E, Liippo J, Lassila O. Quantification of human Aiolos splice variants by real-time PCR. J Immunol Methods 2002; 271:113-23. [PMID: 12445735 DOI: 10.1016/s0022-1759(02)00370-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Aiolos is a transcriptional regulator of B cell development and belongs to the Ikaros family of chromatin remodelling transcription factors. All the members of Ikaros family produce multiple isoforms via alternative mRNA splicing. Altered expression of Ikaros isoforms has been found in patients with acute lymphoblastic leukemia but it is not studied whether the altered expression of Aiolos isoforms also has a role in the development of leukemias or lymphomas. We developed a quantitative real-time PCR application to detect the relative expression of Aiolos splice variants. The method is based on fluorescence resonance energy transfer (FRET)-labelled isoform specific hybridisation probes used with the LightCycler instrument. The isoform specificity is obtained by targeting the probes at the edges of chosen exons. The probes are here shown to represent a rapid, high throughput, specific and reproducible quantification method. We designed and optimised the analysis for a dominant negative Aiolos isoform, but the described method is applicable to any isoform-forming gene. This study shows that the real-time PCR with exon edge spanning probe pairs can be applied generally to reveal the importance of alternative splicing and the role of isoforms in normal development and diseases.
Collapse
Affiliation(s)
- Elli Veistinen
- Department of Medical Microbiology, Turku Graduate School of Biomedical Sciences, University of Turku, Kiinamyllynkatu 13, Finland.
| | | | | |
Collapse
|
43
|
Hübner S, Jans DA, Drenckhahn D. Roles of cytoskeletal and junctional plaque proteins in nuclear signaling. INTERNATIONAL REVIEW OF CYTOLOGY 2002; 208:207-65. [PMID: 11510569 DOI: 10.1016/s0074-7696(01)08005-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Cytoplasmic junctional plaque proteins play an important role at intercellular junctions. They link transmembrane cell adhesion molecules to components of the cytoskeleton, thereby playing an important role in the control of many cellular processes. Recent studies on the subcellular distribution of some plaque proteins have revealed that a number of these proteins are able to localize in the nucleus. This dual location indicates that in addition to promoting adhesive interactions, plaque proteins may also play a direct role in nuclear processes, and in particular in the transfer of signals from the membrane to the nucleus. Therefore, translocation of plaque proteins into the nucleus in response to extracellular signals could represent a novel and direct mechanism by which signals can be transmitted from the plasma membrane to the nucleus. This could allow cells to respond to changing environmental conditions in a rapid and efficient way. In addition, conditional sequestration of karyophilic proteins at the sites of cell-cell and cell-substratum adhesion may represent a general mechanism for the regulation of nucleocytoplasmic transport.
Collapse
Affiliation(s)
- S Hübner
- Institut für Anatomie, Universität Würzburg, Germany
| | | | | |
Collapse
|
44
|
Pérez-Ferreiro CM, Luque CM, Correas I. 4.1R proteins associate with interphase microtubules in human T cells: a 4.1R constitutive region is involved in tubulin binding. J Biol Chem 2001; 276:44785-91. [PMID: 11579097 DOI: 10.1074/jbc.m107369200] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Red blood cell protein 4.1 (4.1R) is an 80-kDa protein that stabilizes the spectrin-actin network and anchors it to the plasma membrane. To contribute to the characterization of functional roles and partners of specific nonerythroid 4.1R isoforms, we analyzed 4.1R in human T cells and found that endogenous 4.1R was distributed to the microtubule network. Transfection experiments of T cell 4.1R cDNAs in conjunction with confocal microscopy analysis revealed the colocalization of exogenous 4.1R isoforms with the tubulin skeleton. Biochemical analyses using Taxol (paclitaxel)-polymerized microtubules from stably transfected T cells confirmed the association of the exogenous 4.1R proteins with microtubules. Consistent with this, endogenous 4.1R immunoreactive proteins were also detected in the microtubule-containing fraction. In vitro binding assays using glutathione S-transferase-4.1R fusion proteins showed that a constitutive domain of the 4.1R molecule, one that is therefore present in all 4.1R isoforms, is responsible for the association with tubulin. A 22-amino acid sequence comprised in this domain and containing heptad repeats of leucine residues was essential for tubulin binding. Furthermore, ectopic expression of 4.1R in COS-7 cells provoked microtubule disorganization. Our results suggest an involvement of 4.1R in interphase microtubule architecture and support the hypothesis that some 4.1R functional activities are cell type-regulated.
Collapse
Affiliation(s)
- C M Pérez-Ferreiro
- Centro de Biologia Molecular "Severo Ochoa," Departamento de Biologia Molecular, Facultad de Ciencias, Universidad Autónoma de Madrid, E-28049 Madrid, Spain
| | | | | |
Collapse
|
45
|
An XL, Takakuwa Y, Manno S, Han BG, Gascard P, Mohandas N. Structural and functional characterization of protein 4.1R-phosphatidylserine interaction: potential role in 4.1R sorting within cells. J Biol Chem 2001; 276:35778-85. [PMID: 11423550 DOI: 10.1074/jbc.m101364200] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Erythrocyte protein 4.1R is a multifunctional protein that binds to various membrane proteins and to phosphatidylserine. In the present study, we report two important observations concerning 4.1R-phosphatidylserine interaction. Biochemically, a major finding of the present study is that 4.1R binding to phosphatidylserine appears to be a two-step process in which 4.1R first interacts with serine head group of phosphatidylserine through the positively charged amino acids YKRS and subsequently forms a tight hydrophobic interaction with fatty acid moieties. 4.1R failed to dissociate from phosphatidylserine liposomes under high ionic strength but could be released specifically by phospholipase A(2) but not by phospholipase C or D. Biochemical analyses showed that acyl chains were associated with 4.1R released by phospholipase A(2). Importantly, the association of acyl chains with 4.1R impaired its ability to interact with calmodulin, band 3, and glycophorin C. Removal of acyl chains restored 4.1R binding. These data indicate that acyl chains of phosphatidylserine play an important role in its interaction with 4.1R and on 4.1R function. In terms of biological significance, we have obtained evidence that 4.1R-phosphatidylserine interaction may play an important role in cellular sorting of 4.1R.
Collapse
Affiliation(s)
- X L An
- Department of Biochemistry, School of Medicine, Tokyo Women's Medical University, Shinjuku, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
46
|
Scott C, Phillips GW, Baines AJ. Properties of the C-terminal domain of 4.1 proteins. EUROPEAN JOURNAL OF BIOCHEMISTRY 2001; 268:3709-17. [PMID: 11432737 DOI: 10.1046/j.1432-1327.2001.02276.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
At the C-terminus of all known 4.1 proteins is a sequence domain unique to these proteins, known as the C-terminal domain (CTD). Mammalian CTDs are associated with a growing number of protein-protein interactions, although such activities have yet to be associated with invertebrate CTDs. Mammalian CTDs are generally defined by sequence alignment as encoded by exons 18-21. Comparison of known vertebrate 4.1 proteins with invertebrate (Caenorhabditis elegans and Drosophila melanogaster) 4.1 proteins indicates that mammalian 4.1 exon 19 represents a vertebrate adaptation that extends the sequence of the CTD with a Ser/Thr-rich sequence. The CTD was first described as a 22/24-kDa domain by chymotryptic digestion of erythrocyte 4.1 (4.1R) [Leto, T.L. & Marchesi, V.T. (1984) J. Biol. Chem. 259, 4603-4608]. Here we show that in 4.1R the 22/24-kDa fragment is not stable but rapidly processed to a 15-kDa fragment by chymotrypsin. The 15-kDa fragment is extremely stable, being resistant to overnight digestion in chymotrypsin on ice. Analysis of this fragment indicates that it is derived from residues 709-858 (SwissProt accession no. P48193), and represents the CTD of 4.1R. The fragment behaves as a globular monomer in solution. Secondary-structure predictions indicate that this domain is composed of five or six beta strands with an alpha helix before the most C-terminal of these. Together these data indicate that the CTD probably represents an independent folding structure which has gained function since the divergence of vertebrates from invertebrates.
Collapse
Affiliation(s)
- C Scott
- Department of Biosciences, University of Kent, Canterbury, Kent, UK
| | | | | |
Collapse
|
47
|
Abstract
Although the mature enucleated erythrocyte is no longer active in nuclear processes such as pre-mRNA splicing, the function of many of its major structural proteins is dependent on alternative splicing choices made during the earlier stages of erythropoiesis. These splicing decisions fundamentally regulate many aspects of protein structure and function by governing the inclusion or exclusion of exons that encode protein interaction domains, regulatory signals, or translation initiation or termination sites. Alternative splicing events may be partially or entirely erythroid-specific, ie, distinct from the splicing patterns imposed on the same transcripts in nonerythroid cells. Moreover, differentiation stage-specific splicing "switches" may alter the structure and function of erythroid proteins in physiologically important ways as the cell is morphologically and functionally remodeled during normal differentiation. Derangements in the splicing of individual mutated pre-mRNAs can produce synthesis of truncated or unstable proteins that are responsible for numerous erythrocyte disorders. This review will summarize the salient features of regulated alternative splicing in general, review existing information concerning the widespread extent of alternative splicing among erythroid genes, and describe recent studies that are beginning to uncover the mechanisms that regulate an erythroid splicing switch in the protein 4.1R gene.
Collapse
Affiliation(s)
- V C Hou
- Lawrence Berkeley National Laboratory, Life Sciences Division, Berkeley, California, USA.
| | | |
Collapse
|
48
|
Mattagajasingh SN, Huang SC, Hartenstein JS, Benz EJ. Characterization of the interaction between protein 4.1R and ZO-2. A possible link between the tight junction and the actin cytoskeleton. J Biol Chem 2000; 275:30573-85. [PMID: 10874042 DOI: 10.1074/jbc.m004578200] [Citation(s) in RCA: 117] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Multiple isoforms of the red cell protein 4.1R are expressed in nonerythroid cells, including novel 135-kDa isoforms. Using a yeast two-hybrid system, immunocolocalization, immunoprecipitation, and in vitro binding studies, we found that two 4.1R isoforms of 135 and 150 kDa specifically interact with the protein ZO-2 (zonula occludens-2). 4.1R is colocalized with ZO-2 and occludin at Madin-Darby canine kidney (MDCK) cell tight junctions. Both isoforms of 4.1R coprecipitated with proteins that organize tight junctions such as ZO-2, ZO-1, and occludin. Western blot analysis also revealed the presence of actin and alpha-spectrin in these immunoprecipitates. Association of 4.1R isoforms with these tight junction and cytoskeletal proteins was found to be specific for the tight junction and was not seen in nonconfluent MDCK cells. The amino acid residues that sustain the interaction between 4.1R and ZO-2 reside within the amino acids encoded by exons 19-21 of 4.1R and residues 1054-1118 of ZO-2. Exogenously expressed 4.1R containing the spectrin/actin- and ZO-2-binding domains was recruited to tight junctions in confluent MDCK cells. Taken together, our results suggest that 4.1R might play an important role in organization and function of the tight junction by establishing a link between the tight junction and the actin cytoskeleton.
Collapse
Affiliation(s)
- S N Mattagajasingh
- Department of Medicine, The Johns Hopkins University School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205, USA
| | | | | | | |
Collapse
|
49
|
Luque CM, Correas I. A constitutive region is responsible for nuclear targeting of 4.1R: modulation by alternative sequences results in differential intracellular localization. J Cell Sci 2000; 113 ( Pt 13):2485-95. [PMID: 10852827 DOI: 10.1242/jcs.113.13.2485] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Red blood cell protein 4.1, 4.1R, is an extreme variation on the theme of isoform multiplicity. The diverse 4.1R isoforms, mainly generated by alternative pre-mRNA splicing, are localized at different intracellular sites, including the nucleus. To characterize nonerythroid 4.1 proteins lacking the most upstream translation initiation site, analyze their intracellular localization and define specific domains involved in differential intracellular targeting of 4.1R, we cloned 4.1 cDNAs lacking that translation initiation site. Seven different 4.1R cDNAs were isolated. Four of these encoded 4.1R proteins localized predominantly to the nucleus and the other three localized to the cytoplasm. Three of the nuclear 4.1R isoforms did not contain the nuclear localization signal previously identified in the alternative exon 16. A comparative analysis of the exon composition of the naturally occurring 4.1R cDNAs cloned and of appropriate composite cDNA constructs, with the subcellular distribution of their respective products, demonstrated that a region encoded by constitutive exons, which is therefore common to all 4.1R isoforms and has been termed ‘core region’, had the capacity of localizing to the nucleus. This region was able to confer nuclear targeting to a cytosolic reporter. In protein 4.1R isoforms, the nuclear targeting of the core region is modulated by the expression of alternative exons. Thus, exon 5-encoded sequences eclipsed nuclear entry of the core region, resulting in 4.1R isoforms that predominantly distributed to the cytoplasm. Exon 5 was also able to confer cytoplasmic localization to a nuclear reporter. In protein 4.1R isoforms, when exons 5 and 16 were both expressed the nuclear targeting effect of exon 16 was dominant to the inhibitory effect observed by the expression of exon 5, yielding proteins that predominantly localized to the nucleus. Taken together, these results indicate that all 4.1R molecules contain a conserved region that is sufficient to target the protein to the nucleus, but that specific exon-encoded sequences modulate this capacity by acting in a hierarchical order.
Collapse
Affiliation(s)
- C M Luque
- Centro de Biología Molecular 'Severo Ochoa' (CSIC/UAM), Universidad Autónoma de Madrid, E-28049 Madrid, Spain.
| | | |
Collapse
|
50
|
Abstract
This article presents new insights into potential roles that three erythrocyte cytoskeletal proteins, protein 4.1, ankyrin, and spectrin, may play in nonerythroid nucleated cells. Each of these proteins is encoded by several closely related genes characterized by complex alternative splicing of their pre-mRNA, thus resulting in the cellular expression of a broad repertoire of isoforms that can adopt tissue- and cell-specific distribution. This could account for the presence of skeletal networks in intracellular organelles such as lysosomes, the Golgi apparatus, or the nucleus. In addition to providing structural support to cell membranes, these cytoskeletal proteins regulate the functions of various transmembrane proteins they interact with, in particular ion channels, as well as the activity of membrane-bound enzymes. Thus, they appear to be key players in major unsuspected cell functions such as protein sorting, dynamics of nuclear architecture during mitosis, or regulation of signal transduction pathways.
Collapse
Affiliation(s)
- P Gascard
- Lawrence Berkeley National Laboratory, Department of Subcellular Structure, California 94720, USA
| | | |
Collapse
|