JD» , Jannfi . 3:... O ..Q ‘3 z . 5.33%qu y 3. I. P» l. A , .. .. n 33.9.53 n V fir? . . . 3r y. «m btmhnufi . an... lung. mi .323183‘3 xv. 51‘ I . hr in”! 5d“... E 1 angina! .I... . .p. 2 Q .oh. {and Jana-mm. . § . 1550.51 I. £ 5...? 93.3.5...» 1.3.... 31);). 11:! >6: z... [.1:!.?:...{ . :55)! ’lxi.‘:. i). .9. 1. .tv I\a ’91.. Mug. .1 . y. ) '1‘) 3.. ‘ rp‘. 2 t I a,\ o, l I THESIS ’HIIIUNHHIHIHIJWI’HIHIIIHllllJUIllll 31293 01417 2252 LIBRARY Michigan State University This is to certify that the dissertation entitled Evidence for an Association Between Splicing Components: Galectin-3 and Polypeptide“) of snRNA presented by SING-YUAN WANG has been accepted towards fulfillment of the requirements for Ph.D. degree in Biochemistry W7wfl Major professor Date 575 HI MS U is an Affirmatiw Action/Equal Opportunity Institution 0-12771 PLACE IN RETURN BOX to remove thle checkout from your record. TO AVOID FINES return on or More dete due. DATE DUE DATE DUE ‘ DATE DUE [—T—l l MSU le An Afflnnetlve Action/Emu Opportunity lnetltution more» i EVIDENCE FOR AN ASSOCIATION BETWEEN SPLICING COMPONENTS: GALECTIN-3 AND POLYPEPTIDE(S) OF snRNP By Sung Yuan Wang A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Biochemistry l 995 ABSTRACT EVIDENCE FOR AN ASSOCIATION BETWEEN SPLICING COMPONENTS: GALECTIN-3 AND POLYPEPTIDE(S) OF snRNP By Sung Yuan Wang Galectin-3 is a galactose/lactose-binding protein found in the nucleus and cytoplasm of a variety of cell types. Using a cell-free assay for pre-mRNA splicing, previous studies have demonstrated that galectin-3 is a required factor for spliceosome formation and splicing activity. Depletion of the lectin from a splicing competent extract resulted in a loss of both activities, which could be restored upon reconstitution with purified recombinant galectin-B. The goal of this thesis was to demonstrate an association of galectin-3 with components of the splicing machinery; specifically, the Sm epitopes found on polypeptides of the small nuclear ribonucleoproteins (snRNPs). This was accomplished at two levels: (a) at the level of single cells, the immunofluorescence patterns of galectin-3 were Shown to be similar to that of the snRNP Sm polypeptides under a variety of conditions and colocalization could be demonstrated in double labeling experiments; and (b) at the level of protein-protein interactions, it was shown that when anti-Mac 2, a monoclonal antibody specific for galectin-B, immunoprecipitates galectin-3 from nuclear extracts of HeLa cells and mouse 3T3 fibroblasts, the Sm B polypeptides are coprecipitated. The immunofluorescence experiments were carried out with mouse 3T3 fibroblasts. The staining patterns for galectin-3 and snRNP Sm polypeptides were monitored in parallel and were found to behave in the same fashion: (a) both yielded diffuse distribution of fluorescence covering the entire nucleus, except for several nucleoli, in fixed and permeabilized cells; (b) using conditions of penneabilization and extraction that preserved a RNP—containing nuclear matrix, both yielded speckled staining patterns; (c) the fluorescence staining was lost for both galectin-3 and snRNP Sm polypeptides when permeabilized nuclear residues were treated with ribonuclease A, whereas neither staining was sensitive to deoxyribonuclease Itreatment; and ((1) during the various stages of mitosis, both galectin-3 and snRNP Sm polypeptides were found to be excluded from the area of chromosomal localization, condensation and separation. Nuclear splicing extracts were subjected to immunoprecipitation with anti-Mac 2. Immunoblotting of the anti-Mac 2 precipitate with polyclonal rabbit anti-galectin-3 revealed the presence of the lectin. Immunoblotting of the same anti-Mac 2 precipitate with an autoimmune serum reactive with the snRNP B polypeptides showed that a fraction of the Sm B polypeptides in the nuclear extract was coprecipitated with the lectin by the monoclonal antibody. The specificity of this coimmunoprecipitation was analyzed by comparing the polypeptides precipitated by anti-Mac 2 versus those found in the corresponding precipitate fractions obtained with anti-Sm (positive control) and with anti- transferrin receptor, an isotype-matched monoclonal antibody (negative control). The coprecipitation of Sm B with galectin-3 by anti-Mac 2 was dependent on the presence of the lectin. Nuclear extracts depleted of galectin-3 by prior adsorption on a lactose affinity resin failed to yield Sm B in the anti-Mac 2 precipitate. The coprecipitation was not affected by the addition of lactose; nor was it perturbed by prior treatment of the nuclear extract with ribonuclease. These results suggest that at least a fraction of the Sm B polypeptides in the splicing extract can interact with galectin-3 through protein-protein interactions. In the course of these studies on the polypeptides precipitated by anti-Mac 2, we also analyzed the composition and identities of polypeptides bound to the affinity resin lactose-agarose under conditions used to deplete the nuclear extract of splicing activity. We found that, in addition to galectin-3, the bound fraction of the lactose-agarose contained at least three other polypeptides. Of particular interest was the identification of one of the other polypeptides to be galectin-1 (the prototype member of the galectin family), whose presence in the nucleus of HeLa cells was confirmed by confocal fluorescence microscopy. These findings provide an explanation for previous observations that while lactose-agarose can deplete nuclear extracts of splicing activity, anti-Mac 2 precipitation failed to yield the same effect. Together with the observation that galectin-1, as well as galectin-3, can alone reconstitute splicing activity in a lactose-agarose depleted extract, these results suggest that the activities of galectin-1 and galectin-3 are redundant in the cell nucleus. To Yi Yu and Catherine and To My Parents for their love, support and faith in me {would iil muggemem d: science and life. i encouragemenl. S in: scientific an. love a s; lit he provided ‘ when of my g hmmmm; Finally. I colleagues Anan ET Amoys. Sh. Emmml ltelugan State. ACKNOWLEDGMENT I would like to express my sincere appreciation to Dr. John L. Wang for his advice and encouragement during my graduate study in his laboratory. I thank him for teaching me about science and life. I would also like to extend my gratitude to Mrs. Patty Voss for her encouragement, support and tolerance. I thank her for providing a conductive environment for both scientific and social growth. I owe a special note of thanks to Dr. Ronald Patterson for his valuable advice and support that he provided me throughout the years. I would also like to express my appreciation to members of my guidance committee, Dr. Richard Anderson, Dr. Zachary Burton and Dr. Arnold Revzin for their advice and criticisms. Finally, I wish to thank Dr. Melvin Schindler, Dr. Siu-Cheong Ho and my friends and colleagues Anandita, Mark Kadrofske, Yeou Guang Tsay, Dr. Jamie Laing, Dr. Neera Agrwal, Eric Amoys, Sharon Grabski who shared both the ups and downs and who made my time in the lab very special. Knowing them has been a privilege and worth by itself the time I have spent at Michigan State. vi LIST Oil LlSl OF I Clltl’lIl LllTRj A l C BIC CIG Slim MATE Cell TABLE OF CONTENTS Page LIST OF TABLES x LIST OF FIGURES xi CHAPTER I: Literature Review ' 1 LITERATURE REVIEW ON GALECTINS 2 A) Carbohydrate Binding Proteins of Animal Cells 2 B) C-Type Lectins 3 C) Galectins 5 1) Structure of Galectins 5 2) Galectin-3: Chimera of Two Domains lO 3) Carbohydrate-Binding Properties of Galectins l2 4) Subcellular Localization of Galectins l4 5) Functions of Galectins l7 LITERATURE REVIEW ON snRNP PARTICLES AND PRE-mRNA SPLICING - l9 A) Structure and composition of snRNP 19 B) Nuclear Localization of the snRNP 29 C) Cytoplasmic Assembly and Nuclear Import of the snRN P 31 D) Non-snRNP Splicing Factors 34 LITERATURE CITED 38 CHAPTER II: Similarities in the Nuclear Matrix Localization of Galectin-3 and Small Nuclear Ribonucleoproteins (snRN P): Evidence from a Comparative Immunofluorescence Analysis 48 SUMMARY 49 INTRODUCTION 5 1 MATERIALS AND METHODS 53 Cell Cultures - 53 Antibodies 53 vii Irnmunofluorescence Microscopy 53 EffeCts of Perrneabilization, Extraction and Nuclease Treatment 54 V RESULTS 56 Irnmunofluorescence Analysis of Galectin-3 S6 Immunofluorescence of Perrneabilized Cells after Salt Extraction or Nuclease Digestion 59 Immlmofluorescence Analysis as a Function of the Cell Cycle 65 DISCUSSION 76 REFERENCES 79 CHAPTER III: Coimmunoprecipitation of Sm Polypeptides of snRN P with Galectin-3 by a Monoclonal Antibodies Directed against the Lectin 82 SUMMARY 83 INTRODUCTION 85 MATERIALS AND METHODS 87 Cell Cultures 87 Antibodies and Affinity Columns 87 Buffers 88 Nuclear Extract Preparation 89 Immunoprecipitation and Lac-depletion of Nuclear Extract 90 Analysis of RNA in Bound Fraction of Immunoprecipitation and Lac- Adsorption 92 Cesium Sulfate Gradient Fractionation and Immunoprecipitation 93 RESULTS 95 Immunoprecipitation of Nuclear Extract of 3T3 Cells by Autoimmune Anti-Sm and by Monoclonal Anti-Mac 2 95 Immunoprecipitation of HeLa Cell Splicing Extracts 102 Effects of Galectin-Depletion on Coimmunoprecipitation 105 Coimmunoprecipitation Experiments in the presence of Lactose and at High Ionic Strength 106 viii DEC flilPl Sl'l ill CH Analysis of the Anti-Mac-2 Precipitate for RNA 1 14 DISCUSSION 121 REFERENCES 126 CHAPTER IV: Nuclear and Cytoplasmic Localization of Galectins: Evidence from Laser Scanning Confocal Microscopy 128 SUMMARY -- 129 INTRODUCTION 130 MATERIALS AND METHODS 133 Antibodies and Affinity Columns 133 Lac-agarose Affinity Adsorption 133 Immunofluorescence Staining and Laser Scanning Confocal Microscopy --------- 134 RESULTS 136 Polypeptides Bound by Lac-agarose 136 Nuclear and Cytoplasmic Localization of Galectin-l: Evidence from Laser Scanning Confocal Microscopy 139 DISCUSSION 144 REFERENCES 147 CHAPTER V: Concluding Statement 149 ix Cllll’ll Tile 1. ( Table I. l Mei 3 Table 6. F LIST OF TABLES Page CHAPTER I: Table 1. Comparison of C-type Lectins and Galectins ....................................................... 4 Table 2. Members of the Galectin Family .......................................................................... 7 Table 3. Previous Names for Galectin-3 ........................................................................... 11 Table 4. Major Mammalian snRNAs ................................................................................ 21 Table 5. Major mammalian snRNPs ................................................................................. 25 Table 6. Proteins of Mammalian snRNPs ......................................................................... 26 LIST OF FIGURES Page CHAPTER I: Figure 1: Schematic diagram illustrating the domain context and organization of the various galectins ............................................................................................ 8 Figure 2: Schematic diagram illustrating the two major steps in pre-mRN A splicing. ............................................................................................................. 20 Figure 3: Schematic diagram delineating the number and order of assembly of the intermediates in spliceosome formation. .......................................................... 23 Figure 4: Schematic diagram illustrating the nuclearcytoplasmic shuttle in the assembly of the RNA and polypeptide components of U1 and U2 snRNPs .............................................................................................................. 33 Figure 5: Schematic diagram summarizing the context and organization of specific domains in SR family of proteins. .................................................................... 35 CHAPTER II: Figure l: Immunofluorescence staining of 3T3 cells after fixation with paraformaldehyde (4%) and permeabilization with Triton X-100 (0.5%). ....... 57 Figure 2: Comparison of the immunofluorescence staining pattern of 3T3 cells. ............ 60 Figure 3: Effect of enzyme treatments on the intranuclear staining pattern for galectin-3 and the Sm antigen of snRNPs ......................................................... 63 Figure 4: Double immunofluorescence staining for galectin-3 and Sm antigen of snRNPs in the same cells. ................................................................................. 66 Figure 5: Immunofluorescence staining patterns of galectin-3 in 3T3 cells as a function of time following serum stimulation. ................................................. 69 Figure 6: Analysis of the immunofluorescence staining pattern of galectin-3 as 3T3 Cells undergo mitosis ......................................................................................... 71 Figure 7: Analysis of the immunofluorescence staining pattern of the Sm antigens as 3T3 cells undergo mitosis. ............................................................................ 74 CHAPTER III: xi Figure 1: Immunoprecipitation of nuclear extracts of 3T3 cells by autoimmune anti-Sm and by monoclonal anti-Mac 2. ........................................................... 96 Figure 2: Profiles of the density and immunoprecipitation patterns of the individual fractions derived from cesium sulfate gradient sedimentation of nucleoplasm. ..................................................................................................... 99 Figure 3: Immunoprecipitation of HeLa cell splicing extracts by autoimmune anti- Sm and by monoclonal anti-Mac 2. ................................................................ 103 Figure 4: The effect of galectin-3 depletion on the immunoprecipitation of Sm B by anti-Mac 2. ................................................................................................. 107 Figure 5: Immunoprecipitation in the presence of lactose (Lac) of HeLa cell splicing extract by autoimmune anti-Sm and by monoclonal anti-Mac 2 ....... 109 Figure 6: Immunoprecipitation under high ionic strength of HeLa cell splicing extract by autoimmune anti-Sm and by monoclonal anti-Mac 2. ................... 1 12 Figure 7: Analysis of the RNA components when HeLa cell splicing extracts are immunoprecipitated by autoimmune anti—Sm and by monoclonal anti- Mac 2. ............................................................................................................. l 15 Figure 8: The effect of RNase treatment on the immunoprecipitation of Sm B by anti-Mac 2. ...................................................................................................... l 19 CHAPTER IV: Figure 1: The composition and identities of polypeptides in the bound fraction of Lac-agarose adsorption. ................................................................................... 137 Figure 2: The subcellular localization of galectin-1 as revealed by laser scanning confocal fluorescence microscopy. ................................................................. 141 xii CHAPTER I Literature Review LITERATURE REVIEW ON GALECTINS A) Carbohydrate Binding Proteins of Animal Cells There are three main classes of proteins involved in protein-carbohydrate interactions (1): a) enzymes that use carbohydrates and glycoconjugates as substrates, such as glycosidases and glycosyl transferases; b) sugar-binding antibodies; and c) the carbohydrate binding protein (CBP) group. Carbohydrate binding proteins are defined as non-enzymatic and non-immune proteins which can selectively bind specific carbohydrate structures and are now referred to as lectins. Ricin, a toxic protein, was documented as the first lectin from a plant source (2). Since the initial identification, lectins have been found in a variety of organisms, in various tissues and cell types. More recently, much attention has been paid to animal lectins. Since carbohydrate structures such as glycoproteins and glycosaminoglycans have been found in the cytoplasm and the nucleus (3), there has been, in turn, greater interest in the intracellular localization of lectins. Animal lectins have been classified based on the nature of their carbohydrate ligands, their involvement in biological processes, their subcellular localization, and their dependence on divalent cations. However, since the primary structures of many lectins have been determined, the shared sequence characteristic is now one of the most useful way to classify lectins. Based on their amino acid sequences, especially those of characteristic carbohydrate-recognition domains (CRD), most animal lectins can be Classified into two major distinct families (4-6). One is the family of C-type lectins which is found extracellularly or associated with plasma membrane and which requires calcium ions for their sugar-binding activity. The other one is the galectin family (7), consisting of soluble, metal-independent, B-galactoside-binding proteins formerly known as S-type or S-Lac lectins (Table 1). B) C-Tyng Lectins The C-type CRDs derive their name from the fact that they require calcium ions for sugar binding activity. The carbohydrates that their CRDs recognize are diverse, including simple carbohydrates such as galactose, N-acetylglucosamine, and complex glycoconjugates such as sialyl Lewis antigens (5). Although calcium ions are required for CRD binding to its ligand, the other domain(s) of C-type lectins is involved in many of the functions of the different proteins. All of the C-type lectins have primary structures typical of extracellular proteins or transmembrane proteins in that they exhibit a signal sequence for entry into the endoplasmic reticulum. C-type animal lectins are found in serum, extracellular matrix and membranes; thus they can be divided into insoluble and soluble groups (6). The insoluble C-type lectin group includes those transmembrane receptors, such as a type-I receptor whose N-terminal region is extracellular and C-terminal region is cytoplasmic. This group includes selectins, mannose receptor and thrombomodulin. The selectins mediate the initial phase of adhesion between leukocytes and endothelia in a weak transient adhesion. A larger number of membrane-integrated C-type lectins are Table 1. Comparison of C-type Lectins and Galectins Pro ierties, 3 .. . r. r a ” .. i. _-_; Galectins . Calcium requirement Yes No Solubility Variable Soluble Cys residues Disulfides Free thiols Cellular localization Extracellular Intracellular Transmembrane Extracellular Carbohydrate specificity Various B-galactosides Ill/:1" categorized into type-H receptors such as hepatocyte receptor and macrophage receptor. A type-II receptor has a cytoplasmic N-terminal region and an extracellular C-terminal domain. Some of the functions ascribed to these receptors include the removal of glycoproteins by endocytosis and phagocytosis. Soluble C-type lectins include collagen- like proteins such as the mannose-binding protein. This type of lectin is composed of an N-terminal collagenous domain and a C-terminal CRD. Other soluble C-type lectins include proteoglycan core proteins, snake venom lectins, etc. C) alectins 1) Structure of Galectins S-type lectins which possess affinity for B-galactosides and show a significant sequence similarity in the carbohydrate-binding site are now renamed as galectins (7). The general designation of the genes encoding galectins is LGALS (lectin, galactoside- binding, soluble), and gene numbering is being kept consistent with the numbering of the proteins; so that LGALSI encodes galectin-1, and LGALSZ encodes galectin-2, etc (7). The CRD of the galectin family is clearly distinguishable from the corresponding CRD of the calcium-dependent C-type lectin (4, 8). Unlike C-type lectins, galectins have been found both inside and outside of cells. They do not depend on cations for carbohydrate binding activity and are isolated usually as soluble proteins. At present, seven galectins in this family have been studied (Table 2). Galectin-l is isolated as a homodimer with monomer molecular weight of ~ 14 kDa. It is abundant in smooth and skeletal muscle, but is also found in many other cell types. Galectin-Z, originally found in human hepatoma, is isolated as a homodimer (9). Galectin-3, previously known as CBP35 was first isolated from mouse 3T3 fibroblasts (10, 11). It consists of a single polypeptide of ~ 33 kDa. Galectin-4 was found as an abundant rat intestinal 36 kDa lectin ( 12). It is homologous to a 32 kDa B-galactoside-binding protein from Caenorhabditis elegans ( l3). Galectin-S was originally isolated from rat lung as a monomer of ~ 18 kDa (14). Galectin-7 is a 15 kDa monomeric protein found in human keratinocytes (15). Galectin-8 was originally cloned from rat liver (16) with a molecular weight of 34 kDa. Unlike galectin-4, which is abundant in the intestine galectin-8 was found in liver, kidney, cardiac muscle, lung and brain. In addition, there are invertebrate lectins whose CRDs fit the consensus sequence of the galectin CRDs but whose subfamily designations has not been decided. These include the sponge (17) and nematode (l3) S-type lectins. As implied by their nomenclature, the structures of all the galectins have at least one carbohydrate recognition domain (CRD), with conserved amino acid sequence between members of the family and homologues of the same member across different species (Figure 1). Thus, galectin-l galectin-2, galectin-5 and galectin-7 each has one CRD, representative of the prototype of the family. Galectin—4 and galectin-8 represent tandem-repeat type with two CRDs in the structure. Each of the CRDs in galectin-4 shows about 25% sequence identity to vertebrate galectin-1, and has carbohydrate- binding activity. Sequence analysis also showed that galectin-8 exhibits about 34% Table 2. Members of the Galectin Family :‘7 ’GaIeCtin H " Subunit StruCture POIypeptideMr Source ChrOmOsome i l Homodimer 14,000 Various species, Human 22 tissues/cell types 2 Homodimer 14,000 Human Human 22 I 3 Monomer 33,000 Various species, Human 13 l tissues/cell types 4 Monomer 36,000 Rat - 1 (C. elegan) 5 Monomer 18,000 Rat Mouse 11 Mouse 4 6 ? ? ? 7 Monomer 15,000 Human Human 19 1 8 Monomer 34,000 Rat - Figure 1: Schematic diagram illustrating the domain context and organization of the various galectins. The carbohydrate recognition domain(s) (CRD) in each galectin is highlighted by the shaded rectangle. Amino acid residues that are conserved among the various galectins are highlighted in the top. The sequence of a nine residue motif that is tandemly repeated (n times, depending on species) giving rise to a proline- and glycine-rich domain in galectin-3 is indicated. ' e e o 0. .................... ............... 0.... .............. a 1 135:} 59' 1' F 7. L I; v ’ . I 1 Galectin-1 ‘ ' 0‘ , . . . . _ .3, . 0 xi f’fif Q {Ie*’ ! ‘”,sz.efx } 4: 140.72.! >3." .me .o;.-.t . :-t-.‘ Galectin-Z ... . . .‘. (y. . .1. ._. . .............................. (pcAvaxxx). Galectin-s 7 VIVV v v’ve ‘vvv ,I. I . I, v‘ . .'.' ~ ,‘,' ' . . i - . .i . ' '.'~‘ ..'.- . 223.7 v'-_ 2‘77 .‘..’ 3.. ,7,‘ .I v. '4 I ' ‘ i A ‘ A ' '. " :5..- l. ....................... 37...: ‘.Va'-.' 7'1 l'.-'.'>' ~~ v’.‘ '4v-ivn. ..... - ...... ‘reel‘g .--‘ ...... A .......... We .xxg.xs '~.q';_ C.n““"“ui§‘ . -------------------------- V... e .'-'.'-‘e'u‘e n‘e'e‘ .......... ................. Dine-enve'v-v' ‘ugfletinjuj zaiééséaizizéaé;E;;;?;i;i:z:i:i::;i§;:;:.i.:1:;..:i::si;é: l Galectin-7 .............................................. J GalectIn-B ............................................... ..................... 10 sequence identity to galectin-4. Galectin-6 has been mentioned (7), but its characterization has yet to be published. 2) Galectin-3: Chimera of Two Domains The galectin—3 group includes proteins originally isolated from various sources which exhibit B-galactoside binding activity and which have been designated different names (Table 3). Genomic Southern blot analysis suggested that there is only one gene coding for the polypeptides of the galectin-3 group. The C-domain of galectin-3 polypeptide contains both hydrophilic and hydrophobic regions, as is characteristic of many globular proteins. This region is about 35% identical with several members of the galectin-1 group, and 14 amino acid residues in this region are highly conserved among galectin-3 and other members of galectin family (Figure 1). Consequently, the C-domain of galectin-3 is assumed to harbor the CRD. In contrast, the N-domain of galectin-3 polypeptide contains neither a highly hydrophilic nor a hydrophobic region. This domain includes a stretch of eight contiguous 9-residue repeat units having the sequence Pro-Gly- Ala-Tyr-Pro-Gly followed by three other amino acid residues. As a result, this stretch of the sequence is characterized by a high proportion of proline and glycine residues (18). Thus, galectin-3 is classified as a chimera—type galectin . Study on the N-terminal domain of galectin-3 showed that it exhibits no apparent B-galactoside-sugar binding activity; in contrast, the C-terminal domain is sufficient for two independent transitions representing the thermal denaturations of the C-domain sugar-binding (19). Differential scanning calorimetry analysis of galectin-3 has yielded Table 3. Previous Names for Galectin-3 Rat, Human, ll . .9} ‘ “.35... Lung, Brain, Kidney Non-classical Dog secretory pathway 88? 31,000 Rat, Human Basophilic leukemia cell IgE binding actixity Mac-2 32,000 Mouse, Human Macrophage Cell surface antigen L-34 34,000 Mouse Fibrosarcoma Cell surface lectin Melanoma metastatic marker CBP35 35,000 Mouse, Human Lung, Fibroblast Intracellular lectin LBP 35,000 Mouse Macrophage Laminin binding activit _- It; "—3 5m 12 (~ 40 0C) and C-domain (~ 55 0C). The binding of lactose to the intact galectin-3 polypeptide and to the C-domain half molecule shifted the 55 °C transition to ~ 65 °C. These results suggest that ligand binding by galectin-3 is accompanied by a conformational change that significantly stabilizes the polypeptide against thermal denaturation. Analysis of the isoelectric point (pI) of E. coli expressed recombinant galectin-3 showed that the pI value of unmodified polypeptide is 8.7, as determined by calculation from the deduced amino acid sequence and by isoelectric focusing gel electrophoresis analysis (20). However, when mouse 3T3 cell extract was subjected to two-dimensional gel electrophoresis and immunoblotting analysis for galectin-3, two spots corresponding to pI values of 8.7 and 8.2 were observed. The pI 8.2 species represents a post- translational modification of the pI 8.7 polypeptide by the addition of a single phosphate group. 3) Carbohydrate-Binding Promrties of Galectins All mammalian galectins recognize the same structural determinant on lactose and related B-galactosides (10, 12, 21-24); however, they share a higher affinity of binding for lactose than that for galactose (25—27). The affinity of galectin-3 for lactose binding was found to be 50-100 times greater than that for galactose. The critical positions of the lactose molecule include the hydroxyls at positions 4 and 6 of Gal and position 3 of Glc, since substitutions at any of these positions resulted in reduced binding activity. 13 Modification of lactose by incorporating an acetamido group at position 2 of glucose moiety to yield N-acetyllactosamine enhanced the binding of galectins. The sequences responsible for carbohydrate binding are mainly encoded by three exons (28-30), for example exons II, III and IV for galectin-l and IV, V and V1 for galectin-3. Most of the residues directly interacting with the carbohydrate ligand are conserved among the galectins and encoded by a single exon. Mutation of some of these residues in exon HI of galectin-1 resulted in impaired carbohydrate binding activity (31, 32). Site-directed mutagenesis on the CRD of galectin-1 showed that the conserved hydrophilic residues, such as His44, Asn46, Arg48, Asn61, Glu71, and Arg73, are important in carbohydrate recognition (32). Deletion of the N-terminal and C-terminal regions of galectin-1 also decreased the carbohydrate-binding affinity indicating that the other two exons may also be important for carbohydrate binding activity (31). Recently, crystallographic studies of galectin-1 have shown that the lectin appears to form a dimer structure. Each of the monomer associates with one N-acetyllactosamine molecule (33, 34). The hydroxyl groups of the galactose molecule in the disaccharide play major roles in its interactions with the protein. The main determinant of the specificity for galectin-1 binding to carbohydrate resides on the hydroxyl at position 4 of the galactose, which interacts with the side chain of Arg48 and His44 through hydrogen bonds. Other residues involved in the carbohydrate binding include Asn46, which interacts with hydroxyls at positions 3 and 4 through a water molecule, and Asn6l with hydroxyl at position 6. The interaction between the side chains of Lys63 and Trp68 also aligns the two residues to interact with the galactose ring. The hydroxyl group of l4 galactose at position 2 shows no interaction with the proteins. The GlcNAc moiety of the disaccharide also interacts with the protein, but the interaction is less extensive than that of the galactose portion. The GlcNAc-protein interactions are mediated through the contact of the hydroxyl at position 3 with Arg73, Arg48 and Glu71. In addition, the N2 position of the N-acetyl group is involved in the interaction with Arg73, Asp54 and His52. This may explain the enhanced binding of galectin to N—acetyllactosamine over that of lactose. A similar result has been found in the study of the crystal structure of N- acetyllactosamine and recombinant galectin-2 complex (35). 4) Subcellular Localization of Galectins Most of the C-type lectins have been found to be localized either in a defined cellular compartment such as membrane-integrated receptors or exported extracellularly as soluble proteins. In contrast, the cellular localization of galectins is somewhat less clear cut. Galectin-l polypeptides have been shown to be intracellular, though reports vary regarding the nuclear and cytoplasmic distribution. Immunofluorescence staining for galectin-1 on cryostat sections from adult chicken kidney (36), calf pancreas (37) have shown that the lectin is localized in cell nuclei as well as cytoplasm. Similarly, immunocytochemical studies in dorsal root ganglion neurons have shown that galectin-1 could be detected both in the nucleus and cytoplasm of the neurons (38). Immunoelectron microscopy has also localized galectin-l in the nucleus of the epidermal cells from the intermediate layer of chick embryonic skin (39). However, immunofluorescence and ultrastructural studies have also led to explicit statements that anti-galectin-l antibodies failed to label the cell nucleus. In a recent study, 15 immunofluorescence staining of Chinese hamster ovary cells by anti-galectin-l antibodies, detected galectin—1 both intracellularly and extracellularly. However galectin- ] was absent from the nucleus (40). Despite the intracellular localization, there is considerable evidence for the extracellular localization of galectin-1, though no typical secretory signal sequence has been found (8). Studies of export of muscle galectin-1 have shown that during myoblast differentiation the lectin becomes concentrated in evaginations of plasma membrane which pinch off or bud off to form labile lectin-rich extracellular vesicles (41, 42). The exported lectin can then interact with carbohydrate on laminin (43) and other extracellular glycoproteins (44). These observations suggest a possible mechanism for lectin export from the cytosol to the extracellular matrix. Galectin-l is also found to be extemalized upon injection of epinephrine without the involvement of secretion vesicle in Xenopus laevis skin cells (45). Similarly, galectin-3 has been found on the cell surface and inside the cell. Most of the galectin-3 was localized intracellularly, especially in the nucleus as shown by immunofluorescence staining, though a small amount of the lectin was detected on the cell surface (46). Subcellular fractionation studies on rat basophilic leukemia cells showed that the majority of the rat galectin-3 is intracellular including in the nucleus (47). Other studies have also shown the intracellular and extracellular localization of galectin-3 in dorsal root ganglion neurons (38, 48), in murine fibrosarcoma cells and human carcinoma HeLa-S3 (49), and mouse macrophages (50). In Baby Hamster Kidney (BHK) cells, the majority of galectin-3 is found in the cytoplasm and small amounts are l6 deposited on the cell surface and substratum (51). In Madin-Darby canine kidney cells the lectin is expressed and secreted at the apical domain of the polarized cells (52), suggesting that apical secretion may be the possible way for galectin—3 extemalization. A wide difference in the level of galectin-3 in various tissues of mice during developmental stages was noted (26). The induction of galectin-3 in mouse macrophages was found to be dependent on the addition of thioglycolate (50). The regulation of expression and subcellular localization of galectin-3 was studied in detail in mouse 3T3 fibroblasts. Galectin-3 was found primarily in the cytoplasm of quiescent cells; however, in proliferating cells, the expression level increased and is predominantly localized in the nucleus (53). In quiescent cells, the cytoplasmic galectin-3 was found to be phosphorylated (pI 8.2). Upon serum stimulation, the synchronized cells showed an increased level of the phosphorylated form (pI 8.2), both in the cytosol and the nucleus. The level of unmodified form (pI 8.7) also increased but could only be found in the nucleus of the stimulated cells (20). The significance and mechanism of the differential expression and localization of the two isoelectric variants of galectin-3 remain unclear. Nevertheless, one study on human colonic tissue specimens has shown that galectin-3 is concentrated in the nuclei of differentiated epithelial cells (54). The expression and nuclear localization changed significantly during the progression from normal mucosa to adenoma to carcinoma; the lectin is absent from the nuclei of carcinoma cells but still localized in the cytoplasm. These observations suggest that the exclusion of galectin-3 from the nucleus may be related to the neoplastic progression of colon cancer. l7 5) Functions of Galectins It has long been suggested that animal lectins might function in modulating cell- cell and cell-extracellular matrix interactions. For example, galectin-l could be involved in modulating transitory adhesions during embryonic cell movement (55), and may also play a role in the elaboration or organization of extracellular matrix components (56). In skeletal muscle, galectin-1 has been shown to either promote or inhibit cell adhesion. One study showed that galectin-1 binds to laminin and inhibits cell-matrix interactions (43); presumably, binding of galectin-1 to polylactosarnine on laminin in myoblasts interferes with laminin recognition by the major laminin receptor integrin 0431. In contrast, a recent report has shown that that galectin-1 binds to laminin and promotes cell adhesion to laminin (57). Nevertheless, these results suggest that galectin-l may have a role in muscle development. Other studies suggested that galectin-l might participate in regulating cell proliferation (58, 59). On the basis of the identification as a laminin-binding protein (see Table 3), it has been proposed that galectin-3, like galectin-l, might also play a role in cell adhesion (60). Some studies suggested that binding of galectin-3 for both IgE and IgE receptor can trigger activation of mast cells and basophils and play a role in inflammation (61). Two glycoproteins named M2BP-l and M2BP-2 were identified from certain colon cancer cell lines by coprecipitation with galectin-3 (62). The interaction is mediated by CRD of galectin-3 and sugar moieties of the glycoproteins. The localization of MZBP-l and M2BP-2 is not clear, but these glycoproteins may play a role in mediating galectin-3 18 function. The exclusion of galectin-3 from the nuclei of carcinoma cells may be related to the neoplastic progression of colon cancer (54). Several lines of evidence suggest that galectin-3 may play a role in pre-mRNA splicing (63, 64). First, immunofluorescence staining and immunoblotting analyses have shown that ribonuclease A (RN ase) treatment of perrneablized cells can remove nuclear galectin-3 while DNase I failed to do so. In addition, fractionation of nucleoplasm derived from Swiss 3T3 cells on a cesium sulfate density gradient revealed galectin-3 in fractions with densities of 1.30 -1.32 g/ml , corresponding to the range of densities reported for hnRNP and snRNP. Finally, the most direct evidence for a role of galectin-3 in pre-mRNA splicing comes from experiments using a cell free assay. The nuclear extracts used for this assay contain galectin-3, as detected by immunoblotting. Addition of saccharides that bind to galectin-3 with high affinity inhibits the cell free splicing. Most persuasively, depletion of galectin-3 by affinity adsorption on a lactose column resulted in a concomitant loss of splicing activity, which could be restored by reconstitution with recombinant galectin-3. LITERATURE REVIEW ON snRNP PARTICLES AND PRE-mRNA SPLICING A) Structure and commsition of snRN P During and after transcription, the pre-mRNA molecules are processed and transported to the cytoplasm where translation occurs. For most of the RNA polymerase II transcripts, this process includes the addition of m7G cap at the 5’-end, polyadenylation at the 3’-end, assembly with proteins to form hnRNP, splicing to remove noncoding introns and ligation of exons, and the exchange of hnRNP proteins for mRN A proteins. The catalytic elements required for pre-mRN A splicing reaction include snRNPs, non-snRNP splicing factors and probably many other factors. The steps in nuclear pre-mRN A splicing have been elucidated and are illustrated in Figure 2 (65). Pre-mRNA splicing proceeds through a two step mechanism involving two separate transesterification reactions. The first step is cleavage at the 5’ splicing site generating free exon 1 by activating the 2’-OH at the branch site producing a lariat RNA; the second step involves another transesterification reaction at the 3’ splice site to join the exons and remove the intron. The catalytic elements, snRNPs, contain uridine-rich small nuclear RNA, a common set of core polypeptides and one or more snRNP-specific proteins. In mammalian cells, there are six major snRNAs (Table 4), named U1 through U6, ranging in size from 107 to 216 nucleotides (66). The major snRNPs are found located in the nucleoplasm, except for U3 which is localized in the nucleolus. Other minor snRNAs have also been characterized; however, this review will focus on the major snRNPs. The 19 20 Figure 2: Schematic diagram illustrating the two major steps in pre-mRNA splicing. Step 1 is a transesterification reaction leading to the formation of a lariat intermediate. Step 2 is another transesterification reaction leading to intron removal and exon joining. This scheme was modified from reference 65. 21 Table 4. Major Mammalian snRNAs AAUUUG GAUUUUUGG AAUUUUUG 22 major snRNAs are transcribed by RNA polymerase II, except for U6 which is transcribed by RNA polymerase H1 (66, 67). Analysis of pre-mRNA intron regions has revealed some consensus sequences to be complementary to conserved internal sequences in U2 snRNA and at the 5’-end of U1 snRNA (68-70). U1 and U2 snRNPs recognize the consensus sequences at the 5’ splice site and branch site in the initial steps in the splicing reaction (Figure 3). In the meantime U4 and U6 snRNAs bind to each other through an extended complementary region to form the U4/U 6 snRNP complex (71) and then the U4/U6 snRNP particle associates with U5 snRNP (72). This tri-snRNP is then bound to the pre-mRNA/U 1/U 2 complex along with other components for intron removed. The snRNPs are either recycled for other splicing events or degraded. Patients with systemic lupus erythematosus (SLE) often possess antibodies against two nuclear antigens called Sm and RNP. Anti-Sm antibodies can immunoprecipitate five major snRNAs, U1, U2, U4, U6 and U5 (Table 5), and minor snRNAs U7, U8, U9, U10, U11 and U12 (73). This was the first indication that different snRNP particles share identical or at least similar proteins. The major epitopes recognized by the anti-Sm class autoantibodies are antigenic determinants on the 28 kDa B and 16 kDa D polypeptides of the snRNP core proteins (Table 6). In addition to these shared proteins, each snRNA is f011nd to be associated with one or more specific polypeptides to form a specific snRNP Particle. The anti-(U1)RNP autoantibodies found commonly in SLE patients recognize determinants on the Ul-specific proteins (73, 74); this antibody reacts mainly with a U1- SPeCific 70 kDa protein, a 33 kDa A and a 22 kDa C protein (75-77) (Table 6). 23 Figure 3: Schematic diagram delineating the number and order of assembly of the intermediates in spliceosome formation. The pre-mRN A substrate contains two exons (El and E2), a 5’-splicing site (S’SS), a 3’ splicing site (3’88), and a branch point (BP). The association of pre-mRNA with hnRNP leads to the formation of the ATP-independent H complex. In the presence of ATP, addition of U1 and U2 snRNPs results in complex A. The addition of a preassembled U4, U5, U6 snRNP complex to complex A results in complex B formation. An ATP-dependent reaction converts complex B to the spliceosome (complex C). After the product mRNA (E1 and E2 joined) is released, the lariat form of the intron is still associated with the snRNPs in a complex. While the snRNPs are recycled, the lariat intron is degraded. This scheme was modified from reference 65. 25 Table 5. Major mammalian snRN P gun-1'.) ._.,. J ~, ,-.~ ,\1 q“. .............................. U1 Core Polypeptides Nucleoplasm Anti-Sm U1 70K Anti-m3G A Anti-(U1)RNP C U2 Core Polypeptides Nucleoplasm Anti-Sm A’ Anti-m3G B” Anti-(U2)RNP U3 ? Nucleolus Anti-(U3)RNP 34 K Anti-m3G ' U4/U6 Core Polypeptides Nucleoplasm Anti-Sm Anti-m3G U5 Core Polypeptides Nucleoplasm Anti-Sm 100 K Anti-m3G 26 Table 6. Proteins of Mammalian snRNPs Anti-Sm Anti-Sm Anti-Sm Anti—Sm 27 With the exception of U6 and the nucleolar snRNP U3, the major snRNPs share a common core of polypeptides, which include B (28 kDa), D’ (18 kDa), D (16 kDa), E (13 kDa), F (12 kDa) and G ( 11 kDa) proteins (Tables 5 and 6). Some of these polypeptides recognize a conserved sequence motif on snRNAs called the Sm binding site (Table 4) and are assembled to form a BzD’zDzEFG-containing snRNP particles (78, 79). In human cells, one B protein is replaced by a B’ protein (80), but the B’ protein is not found in non-primate mammals. Since U6 snRNA is base-paired with U4 snRNA, it could be isolated by antibodies against the core proteins, though it has no Sm binding site (71). At high ionic strength, only the D, E, F and G polypeptides remain associated with the snRNA. These polypeptides are tightly associated and some of the proteins bind to RNA to protect the Sm consensus sequences from RNase digestion (81). A similar RNase protection pattern was also found with particles containing the B protein. These observations suggest that B protein is associated with the snRNP particle through contact with other polypeptides, not by direct binding to snRNA. Consistent with this notion, biochemical studies of the assembly of snRNP particles have demonstrated that D, E, F and G proteins initially assemble to form a 6S particle; the 6S intermediate binds to snRNA followed by the association of two copies of B protein or one copy each of B and B’ proteins (78, 79, 82). Binding of the core proteins to snRN A is directed by a sequence motif of AUnG (n = 2 - 6) (Table 4). This sequence motif is found in the single-stranded region of the 3’ half of all the anti—Sm precipitable snRNAs U1, U2, U4, and US (81). Insertion of this sequence into a single-stranded heterologous RNA has been shown to in ma if S! M l “19’: a.“ in. \ v“ ‘1‘ SF'!‘ .‘Ml 28 induce the assembly of the snRNP core proteins suggesting that this sequence is necessary and sufficient for core protein binding (81). Besides the core polypeptides, Ul snRNP also contains the Ul-specific 70 kDa protein, the 33 kDa A protein, and the 22 kDa C protein (Tables 5 and 6) (83-85). Sequence elements in the stem-loop structure at the 5’-end of U1 RNA are essential for binding of Ul-specific proteins, U1 70 kDa and 33 kDa A (86-88). Purification of U2 snRNP has identified two unique proteins, a 32 kDa methionine-poor A’ protein and a 28 kDa B” protein (84, 85). The U2-specific B” protein has been found to be homologous to the Ul-specific A protein (89, 90). Both A’ and B” proteins have been found to associate with the 3’-end stem-loop region of U2 RNA (91). Although the shared core proteins remain associated with the snRNA under rigorous isolation conditions, the association of the snRNP-specific proteins is sensitive to ionic strength during isolation. For example, there is no specific protein other than core proteins that could be isolated with the U4/U 6 and U5 snRNPs from HeLa cell extracts prepared in 0.5 M NaCl (85). This suggested that the polypeptide composition of snRNP identified may depend on the isolation condition. Protein cross-linking experiments have shown that B and D proteins are directly associated with E, F and G core proteins but not with each other, and the U1 snRNP-specific U1 70 kDa and 33 kDa A polypeptides can be cross-linked to each other, but not to the core proteins (92, 93). These observations suggested that the U1 snRNP- Specific proteins and the core proteins occupy different domains on U] snRN A. 29 B) Nuclear Localization of the snRN P Immunofluorescence localization studies using human autoantibodies (anti-Sm and anti-UlRNP) have shown that the major snRNPs are localized in the nucleoplasm, while U3 is confined to the nucleolus (94, 95). The snRNPs appear to be concentrated in 20-50 nuclear speckles with a diffuse distribution throughout the nucleoplasm, except for nucleoli (96). The diffuse nuclear staining may represent an excess population of snRNPs, or those snRNPs in transit to or from nascent transcripts, or snRNPs in transit to speckles from a cytoplasmic assembly site, and possibly those Sm antigens dissociated from snRNPs. Non-snRNP splicing factors such as SC-35 also have been localized to nuclear speckled structures (97). However, a splicing factor does not necessarily have to be localized to the speckled structure. The protein U2AF (U2 snRNP auxiliary factor) which facilitates U2 snRNP binding to the intron branch site was shown to be distributed throughout the nucleoplasm (98, 99). Fluorescence in situ hybridization and immuno- electron microscopy have revealed a close association between the newly transcribed c- fos RNA and the nuclear speckled structures (100). This result suggested that the nuclear speckled structures observed by immunofluorescence staining for certain snRNPs and non-snRNP splicing factors are indeed the sites for pre-mRNA splicing. Immunoelectron microscopy studies of the distribution of splicing factor SC35 have shown that the speckled staining pattern corresponds to nuclear structures enriched in interchromatin granules and perichromatin fibrils (101, 102). The interchromatin granules with a diameter of 20-25 nm are linked together by thin fibrils (103). It has been shown that after inhibition of RNA polymerase II transcription, a stable population of 30 poly(A)+RNA reorganizes into fewer large interchromatin granule clusters. Along with thereorganization of interchromatin granules, splicing factor SC35 could also be found in these regions (104). Perichromatin fibrils with a diameter of 3-5 mm are found at the peripheral regions of condensed chromatin and dispersed throughout the interchromatin spaces including on the surface of interchromatin granule clusters (103). In contrast to interchromatin granules, perichromatin fibrils could be rapidly labeled with [3H]uridine, suggesting that these fibrils correspond to the structure containing newly transcribed RNA (103). Various components involved in splicing, snRNPs, hnRNP antigens and a non-snRNP splicing factor SC-35, have all been localized to these fibrils (103). Detergent extraction of cells followed by fluorescence in situ hybridization studies have shown that nuclear poly(A)+RNAs are concentrated primarily within several discrete transcript domains which often surround nucleoli in interphase nuclei. Moreover, poly(A)+RNA localization in the cell nucleus is coincident with the Sm containing snRNP (105). Detergent extraction of cells has previously been shown to retain the nuclear matrix and most of the matrix-associated hnRNP (106). Comparison of nuclei with and without detergent extraction demonstrated that specific nuclear RNAs are unambiguously retained upon nuclear matrix preparation (107). Taken together, these results indicate that upon detergent extraction, these RNP molecules remain in the nucleus and suggest that poly(A)+RNA and snRNP may associate tightly with subnuclear structures, and possibly, interchromatin granules and perichromatin fibrils may correspond to these subnuclear structures. These results provide support that these subnuclear structures are involved in pre-mRNA splicing. 31 Previous studies on the distribution of snRNPs have demonstrated that during mitosis, along with the breakdown of the nuclear envelope, the speckled structures observed in the interphase nucleus disappeared and the snRNPs are distributed diffusely throughout the cell except the region for condensed chromatin (108, 109). Immunofluorescence and immunoprecipitation analyses showed that during mitosis the snRNP particles still retain the structure and protein composition of the particles seen during interphase (108). The snRNP particles begin returning to the daughter nuclei after the chromatin begins decondensing in telophase, and they are returned quantitatively to the daughter nuclei during early G1 phase, at which time the speckled structures are reformed. C) anlasmig Assembly and Nuclear Immrt of the snRNP The snRNAs are transcribed as precursors, with several nucleotides longer than the mature nuclear snRNA. These extra nucleotides of newly transcribed snRNA are removed and a 2,2,7-trimethyl-guanosine cap (m3G cap) is added to the snRNA during maturation and snRNP assembly in the cytoplasm (110). The U6 snRNA is transcribed by polymerase III and is capped at the 5’ end by a methyl group (Table 4) (1 l l). The cap structure provides resistance to 5’ exoribonuclease activity ( l 12). Kinetic studies of assembly of snRNP have shown that newly transcribed snRNA appears transiently in the cytoplasm. Analysis of cytoplasm generated from mouse fibroblasts by sucrose gradient centrifugation followed by immunoprecipitation with anti- Sm antibodies has identified a 68 snRNA-free protein intermediate which consists of the 32 D protein and the E, F, and G proteins. This 6S protein intermediate assembles with free snRNA and then with two copies of the B protein to form the snRNP particle. The mature snRNP is then transported back into the nucleus (Figure 4) (79, 80, 82). In contrast to the core polypeptides, some of the U1 and U2 specific proteins are restricted to the nucleus (Figure 4). The binding of core proteins to the snRNA appears necessary for transport of the snRNA into the nucleus. Mutation of the U2 snRNA Sm consensus sequences, preventing the core protein assembly with snRNA, resulted in the accumulation of the snRNA in the cytoplasm; however, removal of sequence responsible for binding of U2 specific protein does not prevent snRNA core protein assembly or nuclear localization of U2 snRNP. These results suggest that the snRNP nuclear localization signal may be generated by the association of core proteins and snRN A. However, nuclear localization of snRNP may also be affected by the snRNA structure. Both mutant U2 snRNA with 3’ extension sequences and mature U2 snRNA could be immunoprecipitated by anti-Sm antibodies; however, mutant snRNA with 3’-end extension could only be found in the cytoplasm (113). Analysis of the 3’-end extension sequence has predicted an interaction between the 3’ extension and the 5’-end stem-loop structure, but this does not prevent binding of core proteins. Similar results have also been obtained for mutant U1 snRN A carrying 3’—end extension sequence (114). These results indicated that core protein binding alone is not the only factor involved in snRNP nuclear import. Proper snRN A structure is also required for snRNP transport into the nucleus. 33 UZSnRNP U1snRNP Figure 4: Schematic diagram illustrating the nuclearcytoplasmic shuttle in the assembly of the RNA and polypeptide components of U1 and U2 snRNPs. The snRNAs transcribed in the nucleus are exported to the cytoplasm, where the core polypeptides B, D, and E-G are added to form snRNPs. Certain U1- specific and U2-specific polypeptides are also assembled on the respective RNPs. These are then translocated into the nucleus, where addition U-specific and U2-specific polypeptides are added. This scheme are modified from reference 79. 34 D) Non-snRNP Splicing Factors A number of proteins essential for splicing activity are not associated with snRNPs. These proteins contain either a serine/arginine-rich (SR) domain or a RNA recognition motif (RRM). In mammalian cells, splicing factors 8035 (115), ASF/SF2 (116), U2AF (117) and U1 70 kDa polypeptide (l 16) contain the RRM and a SR-rich domain (Figure 5). In Drosophila, the products of the transformer (tra) and transformer 2 (tra2) genes (118), which regulate doublesex (dsx) pre-mRN A splicing, also contain the SR-rich domain (119). It has been shown that tra, tra2, and members of the SR family are sufficient to commit dsx pre—mRNA to female-specific splicing, though individual SR proteins differ significantly in their ability to participate in commitment of splicing complex formation. More importantly, the localization signal for nuclear speckled structures may reside in the SR-rich domain (119). A monoclonal antibody which labels mammalian cells in a speckled pattern (120) has been used to identify a family of nuclear phosphoproteins that contain an SR-rich carboxyl terminal domain (121). SDS-PAGE showed that this SR protein family consists of at least five different proteins (20, 30, 40, 55, and 75 kDa). Except for the 20 kDa polypeptide, a repeated sequence that encompasses an RRM is found in all four SR proteins, and each of the four SR proteins individually can complement a splicing-deficient cytoplasmic S-100 extract. The 30 kDa band contains two distinct proteins, SRp30a and SRp30b which have been shown to be ASF/SF2 and SC35, respectively (122). Each individual SR protein initiated splicing of a different pre-mRNA with substrate specificity (123). These observations suggest that 35 _@ RRM @_ Tra 2 RRM ————-- G @—— ASFISF2 —4- RRM PG ——.— SC35 ——@———— RRM1 RRMz RRM3~—— U2AF Figure 5: Schematic diagram summarizing the context and organization of specific domains in SR family of proteins. The RRM box indicates the RNA recognition domain. The relative location of the serine/glycine rich domain (SR) is shown for each protein. PG and G indicate proline/glycine and glycine rich region, respectively. 36 specific SR proteins have distinct and essential roles in pre-mRNA splicing. One possible role is that SR proteins have different specificities for subclasses of pre-mRNAs, like tra and tra2 gene products for Drosophila sex determination, and regulation of the levels of SR proteins in different cell types contributes to the regulation of cell-specific splice choices. The non-snRNP splicing factor U2AF is required for the binding of U2 snRNP to the pre-mRNA intron branch site. Purified U2AF consists of two polypeptides of 35 kDa and 65 kDa. An in vitro splicing assay using cytoplasmic S-100 extract showed that U2AF is an essential splicing factor and all U2AF activity resides in the 65 kDa polypeptide (99). SC35 has been shown to be required for the splicing reaction, especially for the ATP-dependent complex formation (124). In the presence of ATP, SC35 is required for the interaction of U1 snRNP with both the 5’ and 3’ splice sites along with the U2 snRNP binding to the branch site. ASF/SF2 displays two activities in an in vitro splicing assay. It is required for the assembly of the first detectable ATP— dependent splicing complex (116), and is able to switch utilization of alternative 5’ splice sites in a concentration-dependent manner (125). The SR domain in SF2/ASF is not required for alternative site selection, but is necessary for constitutive splicing, and the RRM motif is essential for alternative splicing (126). The alternative splicing activity of ASF/SF2 can be antagonized by hnRNP A] protein (127). Both ASF/SF2 and hnRNP A] have strand annealing activities, which may promote base pairing of snRNAs to the alternative splice sites in the intron. 37 Recently, the SR proteins have been shown to be able to complement splicing activity in reactions depleted of U1 snRNP, but can not restore splicing activity to either U2 snRNP or U4/U6 snRNP-depleted reactions (128). However, the concentration of SR proteins required to complement the U1 snRNP-depleted splicing reaction is about 10- fold greater than that of the endogenous concentration, and the activity of SR proteins to complement the U1 snRNP-depleted splicing reaction is dependent on the particular pre- mRN A. This observation suggests that U1 snRNA is not essential for catalysis of either of the transesterification steps in splicing, and since SR proteins are thought to bind to RNA in a sequence-specific manner, different pre-mRNAs might have differential activity for recognition by SR proteins. 10. 11. 12. Literature Cited Goldstein, I. J. 1979. Carbohydrate-Protein Interaction. American Chemical Society. ACS Symposium Series; 88. Sharon, N ., and Lis, H. ed. 1989. Lectins, Chapman and Hall Ltd., London Hart, G. W. Haltiwanger, T. S., Holt, G. D., and Kelly, W. G. 1989. Ann. Rev. Biochem. 58, 841-874. Drickamer, K. 1988. J. Biol. Chem. 2Q, 9557-9560. Drickamer, K., and Taylor, M. E. 1993. Annu. Rev. Cell Biol. _9_, 237-264. Hirabayashi, J. 1993. Trend in Glycosci. Glycotech. 5, 251-270. Barondes, S. H., Castronovo, V., Cooper, D. N. W., Cummings, R. D., Drickamer, K., Feizi, T., Gitt, M. A., Hirabayashi, J ., Hughes, R. C., Kasai, K., Leffler, H., Liu, F. -T., Lotan, R., Mercurio, A. M., Monsigny, M., Pillai, S., Poiries, F., Raz, A., Rigby, P., Rini, J. M., and Wang, J. L. 1994. Cell 7_6_, 597-598. Hirabayash, J ., and Kasai, K. 1993 Glycobiolgy 3, 297-304. Gitt, M. A., Massa, S., Leffler, H., and Barondes, S. H. 1992. J. Biol. Chem. 261, 10601-10606. Roff, C. F., Rosevear, P. R., Wang, J. L., and Barker, T. 1983. Biochem. J. 2L1, 625-629. Roff, C. F., and Wang, J. L. 1983. J. Biol. Chem. 258, 10657-10663. Oda, Y., Herrmann, J ., Gitt, M. A., Turck, C. W., Burlingame, A. L., Barondes, S. H., and Leffler, H. 1993. J. Biol. Chem. 26_8, 5929-5939. 38 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. 23. 24. 25. 39 Hirabayashi, J., Satoh, M., and Kasai, K. 1992. J. Biol. Chem. _Zfl, 15485-15490. Gitt, M. A., Wiser, M. F., Leffler, H., Herrmann, J. Xia, Y. R., Massa, S. M., Cooper, D. N. W., Lusis, A. J., and Barondes, S. H. 1995. J. Biol. Chem. fl, 5032-5038. Madsen, P., Rasmussen, H. H., Flint, T., Gromov, P., Kruse, T. A., Honore, B., Vorum, H., and Celis, J. E. 1995. J. Biol. Chem. 2E, 5823-5829. Hadari, Y. R., Paz, K., Dekel, R., Mestrovie, T., Accili, D., and Zick, Y. 1995. J. Biol. Chem. ;7_0, 3447-3453. Pfeifer, K., Haasermann, M., Gamulin, V., Bretting, H., Fahrenholz, F., and Muller, W. E. G. 1993. Glycobiology 3, 179-184. Jia, S and Wang, J. L. 1988. J. Biol. Chem. 2_63, 6009-6011. Agrwal, N., Sun, Q., Wang, S. Y., and Wang, J. L. 1993. J. Biol. Chem. 268, 14932-14939. Cowles, E. A., Agrwal, N ., Anderson, R. L., and Wang, J. L. 1990. J. Biol. Chem. 265, 17706-17712. Lee, R, T., Ichikawa, Y., Allen, H. J., and Lee, Y. C. 1990. J. Biol. Chem. 3, 7864-7871. Abbott, W. M., Hounsell, E. F., and Feizi, T. 1988. Biochem. J. _25_2, 283-287. Sato, S., and Hughes, R. C. 1992. J. Biol. Chem. 261, 6983-6990. Knibbs, T. N., Agrwal, N., Wang, J. L., and Goldstein, I. J. 1993. J. Biol. Chem. _fi, 14940-14947. Leffler, H., and Barondes, S. H. 1986. J. Biol. Chem. 261, 101 19-10126. 26. 27. 28.. 29. 30. 31. 32. 33. 34. 35. 36. 37. 38. 4O Crittenden, L. L., Roff, C. F., and Wang, J. L. 1984. Mol. Cell. Biol. 4, 1252- 1259. Sparrow, C. P., Leffler, H., and Barondes, S. H. 1987. J. Biol. Chem. _2_6_5, 7383- 7390. Gitt, M, A., and Barondes, S. H. 1991. Biochemistry 30, 82-89. Gritzmacher, C. A., Mehl, V. S., and Liu, F. T. 1992. Biochemistry 3_1_, 9533- 9538. Rosenberg, I. M., Iyer, R., Cherayil, B., Chiodino, C., and Pillai, S. 1993. J. Biol. Chem. 2_68, 12393-12400. Abbott, W. M., and Feizi, T. 1991, J. Biol. Chem. ;6_6, 5552-5557. Hirabayashi, J., and Kasai, K. 1991. J. Boil. Chem. 2Q, 23648-23653. Liao, D. I., Kapadia, G., Ahmed, H., Vasta, G. R., and Herzberg, O. 1994. Proc. Natl. Acad. Sci. USA 21, 1428-1432. Boune, Y., Bolgiano, B., Liao, D. L., Stecker, G., Cantau, P. Herzberg, 0., Feizi, T., and Cambillau, C. 1994. Str. Biol. 1, 863-870. Lobsanov, Y. D., Gitt, M. A., Leffler, H., Barondes, S. H., and Rini, J. M. 1993. J. Biol. Chem. _2_6_8, 27034-27038. Beyer, E. C., and Barondes, S. H. 1980. J. Supramol. Struct. 13, 219-227. Childs, T. A., and Feizi, T. 1980. Cell Biol. Int. Rep. 4, 775. Hynes, M. A., Gitt, M., Barondes, S. H., Jessell, T. M., and Buck, L. B. 1990. J. Neurosci. _1_Q, 1004- 1013. 41 Akimoto, Y, Kawakami, H., Oda, Y., Obinata, A., Endo, H., Kasai, K., and 39. Hirano, H. 1992. Exp. Cell Res. 1%, 293-304. 40. Cho, M., and Cummings, R. D. 1995. J. Biol. Chem. 23, 5207-5212. 41.. Cooper, K. N. W., and Barondes, S. H. 1990. J. Cell Biol. 1_10, 1681-1691. 42. Harrison, F. L., and Wilson, T. J. G. 1992. J. Cell Sci. 201, 635-646. 43. Cooper, K. N. W., Massa, S. M., and Barondes, S. H. 1991. J. Biol. Chem. _1_1_5, 1437-1448. 44. Cu, M. J., Wang, W. W., Song, W. K., Cooper, D. N. W., and Kaufman, S. J. 1994. J. Cell Sci. M, 175-181. 45. B013, N. C., Roberson, M., Haywood-Reid, P. L., Cerra, R. F., and Barondes, S. H. 1986. J. Cell Biol. E2. 492—499. 46. Moutsatsos, I. K., Davis, J. M., and Wang, J. L. 1986. J. Cell Biol. 192, 447-483. 47. Gritzmacher, C. A., Robertson, M. W., and Liu, F. T. 1988 J. Immunol. 1_1, 2801-2806. 48. Regan, L. J ., Dodd, J ., Barondes, S. H., and Jessell, T. M. 1986. Proc. Natl. Acad. Sci. U. S. A. _8_3, 2248—2252. Raz, A., and Lotan, R. 1987. Cancer Metastasis Rev. 6, 433-452. 49. 50. Cherayil, B. J., Weiner, S. J ., and Pillai. S. 1989. J. Exp. Med. fl, 1959-1972. 51. Sato, 8., Burdett, I., and Hughes, R. C. 1993. Exp. Cell Res. 201, 8-18. 12. Lindstedt, R., Apodaca, G., Barondes, S. H., Mostov, K. E., and Leffler, H. 1992 J. Biol. Chem.l6_8. 11750-11757. 42 53. Moutsatsos, I. K., Wade, M., Schindler, M., and Wang. J. L. 1987. Proc. Natl. Acad. Sci. USA _83, 6452-6456. 54. Lotz, M. M., Andrews, C. W., Jr., Korzelius, C. A., Lee, E. C., Steele, G. D., Jr.,Clarke, A., and Mercurio, A. M. 1993. Proc. Natl. Acad. Sci. USA. 9_(), 3466- 3470. 55. Zalike, 8.15., Thomson, L. W., and Ledsham, I. M. 1987. J. Cell Sci. _8_8,, 483-493. 56. Catt, J. W., and Harrison, F. L. 1985. J. Cell Sci. 3, 347-359. 57. Zhou, Q., and Cummings, R. D. 1993. Arch. Biochem. Biophys. M, 6-17. 58. Yamaoka, L., Ohno, 8., Kawasaki, H., and Suzuki, K. 1991. Biochem. Biophys. Res. Commun. m, 272-279. 59. Wells, V., and Mallucci, L. 1991. Cell 64, 91-97. Woo, H. J., Lotz, M. M., Jung, J. U, and Mercurio, A. M. 1991, J. Biol Chem. 60. 266, 18419-18422. 61. - Frigeri, L. G., Zuberi, T. I., and Liu, F. T. 1993. Biochemistry fl, 7644-7649. 62. Rosenberg, I., Cherayil, B. J ., Isselbacher, K. J., and Pilai, S. 1991. J. Biol. Chem. 266, 18731-18736. Laing, J. G., and Wang. J. L. 1988. Biochemistry 27_, 5329-5334. $4, Dagher, S. R, Wang, J. L., and Patterson, R. J. 1995. Proc. Natl. Acad. Sci. USA 22, 1213-1217. Sharp, P. A. 1994. Cell fl, 805-815. Guthrie, C., and Patterson, B. 1988. Ann. Rev. Genet. 22, 387-419. 43 Kunkel, G. R., Maser, T. L., Calvet, J. P., and Pederson, T. 1986. Proc. Natl. 67. Acad. Sci. USA Q, 8575-8579. 68. Moore, M. J., and Sharp, P. A. 1993. Nature 366, 364-368. 69. lemer, M. R., Boyle, J. A., Mount, S. M., Wolin, S. L., and Steitz, J. A. 1980. Nature E, 220-224. 70. Parker, R., Sillciano, P. G., and Guthrie, C. 1987. Cell 42, 229-239. 71. Hashimoto, C., and Steitz, J. A., 1984 Nucl. Acids Res. fl, 3283-3293. 72. Konarska, M. M., and Sharp, P. A. 1987.Cell Q, 763-774. 73. Lerner, M. R., and Steitz, J. A. 1979. Proc. Natl. Acad. Sci. USA 76, 5495-5499. 74. Williams, D, G. Stocks, M. R., Smith, P. R., and Maini, R. N. 1986. Immunol. 68, 495-500 75. White, P. J., and Hoch, S. O. 1981. Biochem. Biophys. Res. commun. _192, 365- 371. White, P. J., Billings, P. B., and Hoch, S. O. 1982. J. Immunol. 22_8, 2751-2756. 76. Pettersson, 1., Hinterbergwer, M., Mimori, T., Gottlieb, E., and Steitz, J. A. 1984 77. J. Biol. Chem. 2Q. 5907-5914. 78. Feeney, R. J., Sauterer, R, A., Feeney, J. L., and Zieve, G. W. 1989. J. Biol. Chem. 264, 5776-5783. Fisher, D. E., Conner, G. E., Reeves, N. H., Wisniewolski, R., and Blobel, G. 1985. Cell .42. 751-758. Rokeach, L. A., Jannatipour, M., Haselby, J. A., and Hoch, S. O. 1989. J. Biol Chem. 264, 5024-5030. 81. 82. 83. 84. 85. 86. 87. 88. 89. 90. 91. 92. 44 Liautard, J. P., Sri-Widada, J ., Brunel, C., and Jeanteur, P., 1982. J. Mol. Biol. 1Q, 623-643. Sauterer, R. A., Goyal, A., and Zieve, G. W. 1990. J. Biol. Chem. 265, 1048- 1058. Hinterberger, M., Pettersson, I., and Steitz, J. A. 1983 J. Biol. Chem. 2_58_, 2604- 2613. Kinlaw, C. S., Roberson, B. L., and Berget, S. M. 1983. 23. 7181—7189. Bringmann, P., and Luhnnann, R. 1986. EMBO J. 6, 3509-3516. Hamm, K., van Santen, R. A., Spritz, R. A., and Mattaj, I. W. 1988. Mol. Cell. Biol. _8_, 4781-4791. Patton, J. R., and Pederson, T., 1988. Proc. Natl. Acad. Sci. USA 8_5_, 747-751. Patton, J. R., Habets, W., van Venrooij, W., and Pederson, T. 1989. Mol. Cell. Biol. _8_, 3360-3368. Habet, W. J ., Sillekens, P. T. G., Hoet, M. H., Schalken, J. A., Boebroek, A. J ., Leunissen, J. A., van de Ven, W. J. M., and van Venrooij, W. I. 1987. Proc. Natl. Acad. Sci. USA M., 2421-2425. Sillekens, P. T., Habets, W. J ., Beijer, R. P., and van Venrooij, W. I. 1987. EMBO J. _6_, 3841-3848. Mattaj, I. W., and De Robertis, M. 1985. Cell fl, 111-118. Harris, S. G., Hoch, S. 0., and Smith, H. C. 1988. Biochemistry 21, 4595-4600. 93. 94. 95. 96. 97. 98. 99. 100. 101. 102. 103. 104. 105. 106. 45 Luhrmann, R. snRNP Proteins. 1988. in Structure and Function of Major and Minor Small Nuclear Ribonucleoprotein Particles. Bimstiel, M. L. ed. Springer- Verlag, New York. Nyman, U., Halman, H., Hadlaczly, G., Petterson, 1., Sharp, C., and Ringertz, N. R. 1986. J. Cell Biol. fl, 137-144. Spector, D. L., 1984. Biol. Cell. _5_1, 109-112. Spector, D.L. 1990. Proc. Natl. Acad. Sci. USA 22, 147-151. Fu, X. D., Maniatis, T. 1990. Nature 346, 437-441, Carmo-Fonseca, M., Tollervey, D., Pepperkok, R., Barabinok S. M. L., Merdes, A., Brunner, C., Zamore, P. D., Green, M. R., Hurt, E., and Lamond, A. I. 1991.EMBO J. _1_Q, 195-206, Zamore, P. D., and Green, M. R. 1991. EMBO J. m, 207-214, Huang, 8., and Spector, D. L. 1991. Genes Develop. 6, 2288-2302, Spector,D. L., Fu, X. D., and Maniatis, T. EMBO J. 1991.1_Q, 3467-3481, Carmo-Fonseca, M., Pepperkok, T., Carvalho, M. T., and Lamond, A. I. 1992 J. Cell Biol. M. 1—14, Spector, D. L. 1993. Ann. Rev. Cell Biol. 2, 265-313, Huang, 8., Deerinck, T. J ., Ellisman, M. H., and Spector S. L. 1994. J. Cell Biol. 1%, 877-899, Carter, K. C., Taneja, K. L., and Lawrence, J. B. 1991. J. Cell Biol. 115, 1191- 1202. Fey, E. G., Krochmalnic, G., and Penman S. 1986. J. Cell Biol. 202, 1654-1665. 107. 108. 109. 110. 111. 112. 113. 114. 115. 116. 117. 118. 119. 120. 121. 122. 123. 46 Xing, Y., and Lawrence, J. B. 1991. J. Cell Biol. Q2, 1055-1063. Renter, R., Appel, B., Rinke, J ., and Luhnnann, R. 1985. Exp. Cell Res. 15_9, 63- 79. Spector, D. L., and Smith, H. C. 1986. Exp. Cell Res. E3, 87-94. Mattaj, I. W. 1986. Cell 46, 905-911. Singh, R., and Reddy, T., 1989. Proc. Natl. Acad. Sci. USA _86, 8280-8283. Murthy, K. G. K., Park, P., and Manley, J. L. 1991. Nucl. Acids. Res. 12, 2685- 2692. Konings, D. A. M. and Mattaj, I. W. 1987. Exp. Cell Res. 112, 329-339. Neuman De Vegvar, H. E., and Dahlberg, J. E. 1990. Mol. Cell. Biol. m, 3365- 3375. Fu, X. D., and Maniatis, T. 1992. Science A6, 535-538. Krainer, A. R., Mayeda, A., Kozak, K., and Binns, G. 1991. Cell _66, 383-394. Zamore, P. D., Patton, J. G., and Green, M. R. 1992. Nature 355, 609-614. Tian M., and Maniatis, T. 1993. Cell E, 105-114. Li, H., and Bingham, P. M. 1991. Cell 67, 335-342. Roth, M. B., Murphy, G., and Gall, J. G. 1990. J. Cell Biol. 1_11, 2217-2223. Zahler, A. M., Lane, W. S., Stolk, J. A., and Roth, M. B. 1992 Genes Develop. 6, 837-847. Zahler, A. M., Neugebauer, K. M., Lane, W. S., and Roth M. B. 1993. Science @, 219-222. Fu, X. D. 1993. Nature 366, 82-85. 124. 125. 126. 127. 128. 47 Fu, X. D., and Maniatis, T. 1992. Proc. Natl. Acad. Sci. USA 32, 1725-1729. Zuo, P., and Manley, J. L. 1993. EMBO J. _12, 4727-4737. Caceres, J. F., and Krainer, A. R. 1993. EMBO J. 1_2, 4715-4726. Mayeda, A., and Krainer, A. R. 1992. Cell fl, 365-375. Crispino, J. D., Blencowe, B. J ., and Sharp, P. A. 1994. Science E5, 1866-1869. CHAPTER II Similarities in the Nuclear Matrix Localization of Galectin-3 and Small Nuclear Ribonucleoproteins (snRNP): Evidence from a Comparative Immunofluorescence Analysis 48 SUMNIARY Galectin-3 is a galactose/lactose-specific lectin identified as a required factor in pre- mRNA splicing assayed in a cell-free system. In the present study, a comparative analysis of the immunofluorescence staining patterns was carried out using an antiserum specific for galectin-3 and human autoimmune sera reactive against the Sm antigens of small nuclear ribonucleoproteins (snRNP). Using mouse 3T3 fibroblasts, fixed with paraformaldehyde and permeabilized with Triton X-100, both anti-galectin-3 and anti-Sm yielded intense staining covering the entire nucleus, with the exception of ~5 circles devoid of fluorescence. These "black holes" most probably represent the nucleoli within the nucleus. Permeabilization without fixation, followed by extraction with 0.25 M ammonium sulfate prior to fixation and staining yielded speckled patterns for both anti-galectin-3 and anti-Sm. Thus, differences in description of staining patterns, such as diffuse versus speckled, reflect, for the most part, quantitative differences in either the antigen, the antibody or both rather than intrinsic differences in the localization of the two antigens. The nuclear staining was lost for both anti-galectin-3 and anti-Sm when permeabilized 3T3 cells were treated with ribonuclease A while parallel treatment with deoxyribonuclease I failed to yield the same effect. During the mitotic phase of the cell cycle, when chromosomal condensation and nuclear envelope breakdown begins, galectin-3 can be found predominantly around the area of the nucleus but a band along the axis of the chromosomes is totally devoid of galectin-3. Thus, it appears that galectin-3 is found throughout the cell during mitosis except where 49 50 chromosomal DNA is located. Essentially the same results are obtained with anti-Sm staining. These immunofluorescence data suggest that both galectin-3 and the snRNPs are colocalized on the RNP—containing nuclear matrix. This notion is supported by double iminunofluorescence staining, which indicate that there is a direct one-to-one correspondence between the speckled patterns obtained with anti-galectin-3 and with anti- Sm. INTRODUCTION Galectin-3 is a galactose/lactose-specific carbohydrate-binding protein found in both the cytoplasm and nucleus of a wide variety of cells (1). The amino acid sequence of the polypeptide, deduced from the nucleotide sequence of a cDNA clone (2), exhibited structural features, such as tandem repeats of a sequence motif rich in proline and glycine residues, similar to proteins found in the core polypeptides of heterogeneous nuclear ribonucleoprotein complex (hnRNP) and in spliceosomes (3, 4). Using a cell-free assay for pre-mRNA splicing, it was recently demonstrated that galectin-3 constituted a required factor in spliceosome formation and mRNA processing (5). The subcellular localization of several splicing factors has been studied by immunocytochemistry and by in situ hybridization. Immunofluorescence studies using antibodies specific for the small nuclear ribonucleoproteins (snRNP) and non-snRNP splicing factors, such as SC35, have shown that these components are distributed in a speckled pattern in interphase nuclei (see reference 6 for review). In situ hybridization with oligonucleotide probes complementary to the major spliceosomal snRNAs resulted in a similar observation (7, 8). The demonstration of a role for galectin-3 in cell-free pre-mRN A splicing called to question the subnuclear distribution of the protein, relative to the other known components of RNA splicing. Our initial immunofluorescence studies on galectin-3 showed intense staining of the nucleus, covering the entire organelle, and some staining of the cytoplasm (9). Subsequent studies have found varied labeling patterns, ranging from diffuse 51 52 distribution of fluorescence to distinct punctate staining (10, 11). Thus, the present study was undertaken with three main goals in mind. First, we wanted to clarify the issue of the apparent discrepancy in the labeling patterns of splicing factors by a direct comparison between the immunofluorescence yielded by anti-galectin-3 versus an autoimmune antibody reactive against polypeptides of the snRNPs. We found that qualitative differences in the description of the labeling patterns (diffuse versus speckled) may reflect quantitative differences in the antigen, antibody or both. Second, although we had previously reported the sensitivity of the nuclear staining of anti-galectin-3 to ribonuclease treatment (11), implicating anchorage of the protein to a RNA-containing matrix, no direct comparison with a known splicing factor was available. We now report the results of such a study. Finally, we wished to follow the subcellular localization of galectin-3, along with a known splicing factor, through the entire cell cycle. In the present study, we found that both galectin—3 and snRNPs were excluded from the segregating chromosomes during mitosis. Combined with the biochemical data to be reported in Chapter III, the various lines of evidence strongly suggest that galectin-3 is associated with the storage/assembly sites of splicing factors. MATERIALS AND METHODS Q11 Culture. Mouse 3T3 fibroblasts were obtained from American Type Culture Collection (Rockville, MD). The cells were grown as monolayers in Dulbecco modified Eagle's Medium containing 10% calf serum, 100 U/ml penicillin and 100 ug/ml streptomycin at 37 0C in a humidified atmosphere of 10% C02. Cells grown at a density less than 5 x 104 cells/cm2 were proliferative (sparse culture); above this density, the cells were confluent. In some experiments, cells at low density were arrested by removal of serum and maintenance in medium containing 0.2% calf serum for 48 hours (quiescent cells). Upon readdition of serum (10%), the cells were reactivated (10). Antibodies. In the present study, antiserum against galectin-3 was derived from rabbit #33. This Flemish Giant rabbit was immunized with recombinant galectin-3, purified from an E. coli expression system. The details for the production of recombinant galectin- 3, the generation of the antisera, and the characterization of its specificity have been previously reported (12). Antiserum directed against RAP30 ( 13) was a gift of Dr. Zachary Burton (Michigan State University). Human autoimmune serum reactive with the Sm antigens of snRNPs (ENA anti-Sm) was purchased from The Binding Site (San Diego, CA). Immunofluorescence Microscopy. Mouse 3T3 fibroblasts were seeded onto coverslips (22 x 22 cm), which were then placed in 6 -well (8 cmzlwell) cluster dishes. The cells were washed twice (4 mee11) with phosphate-buffered saline (PBS, 140 mM NaCl, 2.68 mM KCl, 10 mM NazHPO4, 1.47 mM KH2P04, pH 7.4). Cells were fixed by incubation (20 minutes at room temperature) in 4% paraformaldehyde in PBS (2 ml/well), 53 54 followed by washing twice with PBS (10 minutes each, on a rotating platform). The residual aldehyde groups were blocked by 0.1 M glycine in PBS (4 ml/well; 20 minutes at roOrn temperature). After removal of the glycine-PBS solution, cells were permeabilized with 0.5% Triton X-100 in PBS (2 mee11; 4 minutes at room temperature). The cells were again washed twice with PBS. The cells were then incubated with antiserum, with appropriate dilution, for one hour at room temperature. After washing three times in T-TBS (10 mM Tris pH 7.5, 500 mM NaCl, 0.05% Tween 20) (15 minutes each), the cells were incubated with the secondary antibody, at appropriate dilution. Fluorescein-conjugated or rhodamine-conjugated goat anti-rabbit immunoglobulin (BMB, Indianapolis, IN) was used to detect rabbit anti-galectin-3 and rabbit anti-RAP30 binding and fluorescein-conjugated or rhodamine-conjugated goat anti-human immunoglobulin was used to detect human autoimmune ENA anti-Sm binding. After immunostaining, cells were counterstained for DNA with 4',6-diamidino-2-phenylindole-2-HC1 (DAPI) at a concentration of 1 ug/ml (14). Finally, the coverslips were washed three times in T-TBS (15 minutes each) before mounting with Penna-Fluor on glass microscope slides. Effects of Permeabilization Extraction and Nuclease Treatment. In some experiments, 3T3 cells were permeabilized (0.5% Triton X-100 in 300 mM sucrose, 100 mM NaCl, 3 mM MgC12, 10 mM 1,4-piperazinediethanesulfonic acid, pH 7.4; 10 minutes at 4 oC) without prior fixation. The permeabilized cells were washed with PBS twice (30 seconds each) and were either extracted with 0.25 M ammonium sulfate for 10 min at room temperature (15) or were digested with ribonuclease A (RNase; Sigma; 100 ug/ml) or deoxyribonuclease I (DNase; BMB; 100 U/ml) at 37 °C for 1 hour. The treated cells were 55 washed with PBS twice (30 seconds each), and cells were fixed by incubation (20 minutes at room temperature) in 4% paraformaldehyde in PBS (2 ml/well), followed by washing twice with PBS (10 minutes each, on a rotating platform). The residual aldehyde groups were blocked by 0.1 M glycine in PBS (4 mee11; 20 minutes at room temperature). After removal of the glycine-PBS solution, the cells were then stained with antiserum as described above. RESULTS Immunofluorescence Analysis of Galectin-3. We had previously reported the development of polyclonal rabbit antisera against recombinant galectin-3, purified from an E. coli expression system (12). One antiserum (#33) and its preimmune control serum were used in experiments throughout this entire study. This antiserum immunoblots a single polypeptide (Mr 33,000) in extracts of mouse 3T3 fibroblasts. Moreover, immunoblotting analysis of purified NHz- and COOH-tenninal domains of galectin-3 indicated that the principal epitopes recognized by anti-serum #33 were localized within the NHz-terminal half of the polypeptide (12). These results obviate any complication in the interpretation of the staining patterns, particularly in terms of the possibility that the antibody reagent might cross-react with the carbohydrate recognition domain of the members of the galectin family, which exhibit sequence homology with each other (1). Mouse 3T3 fibroblasts, fixed with paraformaldehyde and permeabilized with Triton X- 100, yielded both nuclear and cytoplasmic staining with anti-galectin-3. This was observed both in low magnification micrographs showing a field containing several cells (Figure 1A) as well as high magnification micrographs showing a single cell (Figure 1B). Parallel analysis with the corresponding preimmune serum showed negligible staining (Figure 1E and Figure 1F). In general, the staining appeared to be diffuse, covering the entire nucleus with the exception of ~5 circles devoid of fluorescence (Figure 1B). These "black holes" were also observed when the same cells are counterstained with the DNA- 56 57 Figure 1: Immunofluorescence staining of 3T3 cells after fixation with paraformaldehyde (4%) and permeabilization with Triton X-100 (0.5%). A, E: low magnification showing fields containing several cells; B-D, F-H: high magnification showing single cells. A and B, rabbit anti-galectin-3 (1:150 dilution of antiserum #33); E and F, preimmune serum from the same rabbit #33 (1:150 dilution). The binding of the primary antibody was detected by rhodamine-conjugated goat anti-rabbit immunoglobulin. The cells in B and F are shown in DAPI counterstaining and phase contrast micrographs in C, G and D, H, respectively. (Bar, 50 um) 58 59 specific dye DAPI (Figure 1C and Figure 1G). In phase contrast micrographs of the same cells, these "black holes" correspond to phase dense structures in the nucleus (Figure 1D and Figure 1H). Moreover, the "black holes" were seen in cells treated with antibodies directed against two other nuclear proteins:(a) RAP30, a 30 kDa component of the general transcription factor, TFIIF; and (b) Sm, an epitope defined on proteins B and D of snRNPs (Figure 2, column A). On the basis of these results and comparison with previously published staining patterns of snRNPs and the non-snRNP spliceosome component SC35 (14, 16), we believe the "black holes" correspond to nucleoli. Thus, the nuclear distribution of galectin-3 appears to be diffuse throughout the nucleoplasm, excluding the nucleoli. Immunofluorescence of Permeabilized Cells after Salt Extraction or Nuclease Digestion. When 3T3 cells were permeabilized with Triton X-100 (without fixation), then fixed and stained with antibodies against galectin-3, Sm, and RAP30, the labeling patterns (Figure 2, column B) were qualitatively different from those obtained with cells that were initially fixed prior to permeabilization (Figure 2, column A). First, cytoplasmic staining was lost from the galectin-3 and RAP30 samples. Second, there was reduction in the staining intensity in all three samples. Finally, there appeared to be a general smearing of stained structures, yielding a diffuse distribution of fluorescence and accentuating the nucleolar "black holes" (galectin-3 and Sm). The most striking labeling patterns were obtained, however, when the permeabilized cells were extracted with 0.25 M ammonium sulfate prior to fixation and staining. This procedure extracts the majority of the nonhistone nuclear proteins, leaving chromatin, nuclear matrix, and associated RNAs (15, 17). In all A81 On .38 832030888 888-83 “mow 339.806.8888.... .3 co 8.3238288 838-85 “new 33w3809088meofi .3 388% 83 .3088“ 38.5 05 .8 9583 05. 888% can” 3 838a (95 888 0888685 58:: new .8826 On _ u C 83.85 838 Anne 88883 co noes—6 o2 ” C Wazoo—«mean :38 5:5 3:338 :05 83 cocoa—U8 some 898 0:28.. 2F .82.: :05 28 63:8 83:68.5 2 mmd 55 3885 .38388& 803 8:8 CV 696 :05 was 85 38.538ch 803 2.8 EC 5888888 came 87x :28. 2 38 see 8283888 53 use 2»; £00 93 8:3 nah he F538. 9883» 3:888:=:==EE_ 05 .8 85.—38:0 "N 953m is” 61 Sm 62 cases except RAP30 (which has lost all of its staining), distinct speckled patterns were observed (Figure 2, column C). It should be noted that differences between the diffuse staining pattern seen in column A of Figure 2 and the speckled pattern seen in column C of the same figure (for galectin-3 and Sm) probably reflect, at least in part, quantitative differences in either the antigen, the antibody, or both. With high titer antiserum and high levels of antigen (e.g. in fixed and permeabilized cells of column A), the staining intensity is so strong that it covers the entire nucleus, obscuring even the nucleolar "black holes" (Figure 2, column A, anti- galectin-3 staining). With loss of antigen from the nuclei during the permeabilization and extraction procedures, the staining of discrete structures/regions becomes more distinct, giving rise to a speckled pattern (Figure 2, column C, anti-galectin-3 staining). Alternatively, the same staining protocol could yield, with two different antisera against the same antigen, either diffuse or speckled patterns. In this connection, we have also observed variations in the DAPI staining of DNA, ranging from a completely fluorescent nucleus, diffuse fluorescence except for nucleolar "black holes", to punctate patterns (see, for example, DAPI staining in Figure 3 below). The speckled staining pattern observed for galectin-3 and Sm (Figure 2, column C) suggests that these antigens are associated with subnuclear structures. Therefore, the effects of RNase and DNase on the immunofluorescence patterns of Triton X-100 permeabilized cells (Figure 2, column B) were studied. RNase digestion resulted in drastic reduction in the intensity of galectin-3 staining (Figure 3, columns A and B). There appeared to be some 63 A81 0m .38 823030888 888-88 “mow vouawsmcoerEaEE 80 8:50—38:88 8598-88" “mow Bemw=809088m00€ .3 080200 83 because 3.8.8 05 .80 9583 2; #75 8_3 808882800 08* 80:28 cowH 3 8m85 0 8m-:80 .«0 8000: 8:203 05 0305. m _oc0m 280030888: 2000380 80» 02088092000800: .3 00.032 6-2828280 .0 Essa 0550; 20 $2... < .28 .32 588:5 .o .5020 02 8 0-5.8.0055 fine 3 832.8 828:0 com” S 8m-0:0 €00,823. 0:00 .5405 .23 w0_0m08020000 .3 0032.8 003 <20 3080000000 .«0 00280 05. 020003000005 0300-000 80m 0203.88.05.00005 .3 080800 003 3000000 b00000 05 .8 m0m003 05. .390 8008000 «0 025:0 om _ H: 9000208000 03000 :03 00000005 000 0002302500 .0050 803 0000—0050 05.8 800 0000: en 000 cm 0003003 0003—00 05.0 0:00 600300 000000: 0:00 mph 00 Wage—0w *0 000300 05550 00000000033055“ 2: no $920000 "0 0.53..— ‘? E 33 a U 72 DNA H 73 revealed by DAPI staining, appears as a dark band in the immunofluorescence photograph (Figure 6B). As the chromosomes separate in anaphase (Figure 6C), and decondense in telophase (Figure 6D), galectin-3 is found throughout the cell except where chromosomal DNA is located. As the nuclear envelope reforms around the decondensing chromosomes and the cell undergoes cytokinesis (Figure 6E), galectin-3 appears to be excluded from the nucleus. This cytoplasmic localization of galectin-3 persists in the newly formed daughter cells (Figure 6F). Thus, galectin-3 must be retranslocated into the nucleus as the cells enter the 61 phase of the next cell cycle and cells from cultures 32 hours post serum stimulation yielded staining patterns similar to those from cultures 8 hours after serum addition (Figure 5). Essentially the same results are observed when the Sm antigen of snRNPs are stained with autoimmune serum (Figure 7). The prophase through cytokinesis stages of mitosis are again discerned from DAPI staining of DNA. Like galectin-3, the staining of Sm was excluded from the position of chromosomes. After separation of the daughter cells, Sm was found throughout the cytoplasm and must be retransported into the nucleus. 74 .50: 300000200 0000000 000:: .0000 03000 0.00 0:00 0.000 000. 0:00 0003000 00:00.: .2300 030 Pb 000 ”00000—003 Amy ”000000.00 ADV ”00000000 6V ”0000000000 av 0000:0000 $3 ”03000 80 080000 00 00w000 00000> 300000.500 0:00 .720 0:3 330000000000 3 0032—8 003 <20 00000080000 .00 0000000 05. 00000—300005 000000-000 000w 0000w0.n000.00_E000€ .3 00000000 003 3000000 b00000 0.0 00 w0_005 00,—. 300.50 com” S Em-000 2 883.52”. A3 .wfizofiocsEE .8 Pap—mega .3 332—8 .AoEEEcCoa bend: m0» $8.58 05 98 U Stan E 2.8 30: E0: 3835 203 38.88 wEBEm .N $2.35 an m Em he :o:8_&uo.ae:=EE_ 2: :e Ease—now naive—aw ue «coho 2; 3 ensure 108 109 .Uofiwznwfi 9a fiasco—mm mm :03 an .O Em .m Em mo :23qu no mcoEmom 2F .BSEBBQ N 82-35 “8 AB 28 63:98:“ Eméqm 5:5: Avv U§§8a Ehéca “8 GS "omoawaéj mo 839$ wagon NV 39253 mag—am =8 30: CV “803 0:3 nomo E 8.95% A6 new AS £258 5 .Emé—a 95% of the galectin-3 polypeptide had been removed from the extract. Moreover, the splicing activity of the galectin-3 depleted extract, derived from the anti-Mac 2 adsorption, was still sensitive to inhibition by Lac. This suggests that other Lac- binding proteins may be present in the splicing extract and they can be depleted, along with galectin-3, by Lac-agarose but not by anti-Mac 2. Thus, it seemed important to define the components that are bound by the Lac-agarose column and to test the role of each or combination of these components in terms of the reconstitution of splicing activity. In the meantime, I had been attempting to test the hypothesis, by coimmunoprecipitation studies reported in Chapter III, that galectin-3 interacted with components of the spliceosome machinery. In comparing the polypeptides found in the bound fractions of anti-Mac 2, anti-TfR, anti-Sm, and Lac-agarose precipitations (see for example, Figure 6, Chapter III), it became apparent that at least two other polypeptides, besides galectin-3, was adsorbed by the Lac affinity resin. Thus, it seemed possible to provide an explanation to Sue Dagher's observations (9) if we could definitively identify 132 these polypeptides as Lac-binding proteins and document their nuclear localization. The results of these studies are now reported in this chapter. 133 MATERIALS AND METHODS Antibodies and Aff'mity columns. Polyclonal rabbit anti-galectin-3 antiserum was obtained from rabbit #33 (10). Polyclonal rabbit anti-galectin-l antibody was a gift of Dr. S. H. Barondes (University of California, San Francisco). Rabbit antiserum against lactose dehydrogenase (LDH) was a gift from Dr. John Wilson (Michigan State University). The derivatization and characterization of rabbit anti-calf thymus actin have been reported (11, 12). ENA anti-Sm antibody was purchased from The Binding Site (San Diego, CA). Lactose immobilized on 6% beaded agarose (Lac-agarose) was purchased from Sigma (St. Louis, Mo). Sepharose CL4B beads was obtained from Pierce (Rockford, IL) Lac-agarose Affinity Adsorption. Nuclear extract was prepared according to Dignam et. al.(15) as described in Chapter 3. The [358]methionine labeled nuclear extract (in buffer C, 20 mM HEPES pH 7.9, 25% (v/v) glycerol, 0.42 M NaCl, 1.5 mM MgC12, 0.2 mM EDTA, 0.5 mM PMSF, 0.5 mM DTT) was precleared by incubating with Sepharose beads for 1 hour at 4 0C. The unbound fraction of this preclearing step was subjected to Lac-agarose adsorption. Precleared nuclear extract (50 111) was incubated with 50 pl of Lac-agarose beads (pre-washed with buffer C twice) for 1 hour at 4 0C. The Lac-agarose were washed: (i) twice with buffer 1 (10 mM Tris, pH 7.5, 150 mM NaCl, 0.5% NP-40, 0.2 mM EDTA); (ii) twice with buffer 2 (10 mM Tris, pH 7.5, 500 mM NaCl, 0.5% NP-40, 0.2 mM EDTA); and (iii) twice with buffer 3 (10 mM Tris, pH 7.5). The material remaining bound to the beads were subjected to SDS-PAGE (12.5% 134 acrylamide) as detailed in Chapter 3 and the proteins were revealed by fluorography and by immunoblotting. For immunoblotting analysis, rabbit anti-galectin-3, anti—galectin-l and anti-actin were used at 1:500 dilution. All three primary antibodies were diluted in TBS (10 mM Tris, 0.5 M NaCl, pH 7.5) containing 0.2% gelatin. Horseradish peroxidase conjugated sheep anti-rabbit antibodies (BMB) were used at 1:2,000 dilution in T-TBS (TBS containing 0.05% Tween 20) as secondary antibody. Proteins resolved on a SDS-PAGE were electrophoretically transferred to Immobilon-P (Millipore) membrane as described in Chapter III. The membrane was briefly rinsed twice with T-TBS and blocked in 2% gelatin/TBS for at least 1 hour at room temperature. After briefly washing with T-TBS twice, the membrane was incubated with primary antibody for 1 hour at room temperature. The membrane was then rinsed with T-TBS twice and washed extensively with T-TBS (15 minutes once, then 10 minutes 4 times). The washed membrane was incubated with horseradish peroxidase conjugated secondary antibody at room temperature for 1 hour. The blot was rinsed and washed as described above, and then developed with 4-chloro-1-naphthol. Immunofluorescence Staining and Laser Scanning Confocal Microscopy. HeLa cells grown as monolayers were obtained from American Type Culture Collection (Rockville, MD). They were seeded onto coverslips (22 x 22 cm) in 6-well (8 cm2/well) cluster dishes. The cells were washed twice (4 ml/well) with phosphate-buffered saline (PBS, 0.14 M NaCl, 2.68 mM KCl, 10 mM NazHPO4, 1.47 mM KH2P04, pH 7.4). Cells were fixed by incubation (20 minutes at room temperature) in 4% paraformaldehyde in PBS 135 (2 ml/well), followed by washing twice with PBS (10 minutes each, on a rotating platform). The residual aldehyde groups were blocked by 0.1 M glycine in PBS (4 ml/well; 20 minutes at room temperature). After removal of the glycine—PBS solution, cells were permeabilized with 0.5% Triton X-100 in PBS (2 mee11; 4 minutes at room temperature). The cells were again washed twice with PBS. The cells were then incubated with antiserum (anti-galectin- 3, 1:150; anti-galectin-l, 1:150; anti-LDH, 1:150; and ENA anti—Sm, 1:500) for one hour at room temperature. After washing three times in T-TBS (10 mM Tris pH 7.5, 0.5 M NaCl, 0.05% Tween 20) (15 minutes each), the cells were incubated with the secondary antibody for one hour. Fluorescein-conjugated goat anti-rabbit immunoglobulin (BMB) (1 :50) was used to detect rabbit anti-galectin-3, rabbit anti-galectin-l , and rabbit anti-LDH binding and fluorescein-conjugated goat anti-human immunoglobulin (BMB) (1:100) was used to detect human autoimmune ENA anti-Sm binding. Finally, the coverslips were washed three times in T-TBS (15 minutes each) before mounting with Penna-Fluor on glass microscope slides. A Meridian Instruments (Okemos, MI) Insight bilateral laser scanning confocal microscope was used with an argon ion laser as the excitation source. An 100 x objective lens and laser power of 50 mWatts were used for scanning the image. One photomultiplier was used to detect fluorescence emission of fluorescein (530 nm). Images (360 x 360 pixels) were collected from 9 consecutive focal planes as with an increment of 0.5 pm for each step in the x and y directions (16). RESULTS Polypeptides Bound by Lac-agarose. Nuclear extracts were prepared from HeLa cells metabolically labeled with [3SS]methionine. The extract was "precleared" by sepharose beads to remove components that would bind nonspecifically to the beads. The unbound fraction of the "preclearing" step was then subjected to binding with Lac-agarose. The conditions for these experiments (buffer C) were identical to those used to deplete splicing extracts of galectins and splicing activity (8, 9). The material bound to the beads was subjected to SDS-PAGE, followed by fluorography and immunoblotting. Five polypeptides were found in the bound fraction of the Lac-agarose adsorption with molecular weights of approximately: (a) 70,000; (b) 50,000; (c) 43,000; (d) 30,000; and (e) 15,000 (Figure 1, lane 1). Three of these bands have been identified on the basis of immunoblotting analysis. First, a rabbit anti-calf thymus actin antiserum (reactive predominantly with non-muscle actins (13, 14)) immunoblotted the 43 kDa band (Figure 1, lane 4), suggesting it to be actin. This would be consistent with the observation that this polypeptide was also bound to the preclearing sepharose beads in a nonspecific fashion (see, for example, Figure 6, Chapter 111). Second, polyclonal rabbit anti-galectin-3 identified the 29 kDa band as galectin-3 (Figure 1, lane 3), as was reported previously (8) that under these conditions (high ionic strength of buffer C), Lac-agarose depleted splicing extracts of galectin-3 with concomitant loss of splicing activity. Finally, the 14 kDa band was identified as galectin-1, on the basis 136 137 Figure 1: The composition and identities of polypeptides in the bound fraction of Lac-agarose adsorption. Nuclear extracts were prepared, following the procedures of Dignam et a1. ( 15), from HeLa cells metabolically labeled with [35$]methionine. The nuclear extracts, in buffer C, were subjected to affinity adsorption on Lac-agarose beads. The bound fraction was then subjected to SDS-PAGE ( 12.5% acrylamide), followed by fluorography or by immunoblotting with various antibodies. Lane 1: Radioactive polypeptides revealed by fluorography. Lane 2: Irnmunoblot with anti-galectin-l. Lane 3: Irnmunoblot with anti-galectin-B. Lane 4: Irnmunoblot with anti-actin. The positions of migration of authentic protein markers are highlighted on the left. tlmrllncfin es of Dignam cl . mine. 111cm: on on lac-aging: 125% acryltrai arious mimic) Lane 2: Irrrmrttt1 actin-3. land: 1 of authenticgm 138 139 of immunoblotting with affinity purified anti-galectin-l antibodies (Figure 1, lane 2). Thus, it appears that Lac-agarose was indeed depleting more than one Lac-binding protein from the nuclear extract (i.e. galectin-1, in addition to galectin-3). The identities of the other prominent band (Mr 50,000) and minor band (Mr 70,000) are not known. That the prominent band represents a bona fide Lac-binding protein is suggested by preliminary experiments showing its specific elution from the Lac-agarose beads by the addition of Lac. Neither of these two bands reacted with any of the anti- galectin antibodies available to us, including an antiserum against rat intestinal galectin-4. Nuclear and Cytoplasmic Localization of Galectin-l: Evidence from Laser Scanning Confocal Microscopy. The identification of galectin-1 in nuclear extracts of HeLa cells and its adsorption onto Lac affinity resins provided an explanation to the observation that while Lac-agarose can deplete the splicing activity, anti-Mac 2, a monoclonal antibody specifically reactive with galectin-3, failed to do so. Coupled with the demonstration that either galectin-l or galectin-3 alone is sufficient to reconstitute splicing activity in a Lac-agarose depleted extract (9), the results would suggest that the activities of galectin-1 and galectin-3 in the nucleus may be redundant. On the other hand, however, the notion that galectin-1 can be found in the nucleus is controversial, particularly since, in the original publication reporting the purification of the chicken homologue, there was an explicit statement indicating that anti-chicken heart galectin-1 failed to label the cell nucleus of chicken embryo fibroblasts under immunofluorescence (17). The main difficulty in the analysis of galectin-1 is that, unlike galectin-3, the predominant portion of the former lectin is found in the cytoplasm. As a result, a strong fluorescent antibody probe staining the 140 cytbplasm surrounding the nucleus sometimes results in an image containing fluorescent flare that exceeds the resolution of the light microscope. To circumvent this difficulty, we took advantage of the availability of a laser scanning confocal microscope to analyze the staining pattern obtained with anti-galectin-l. HeLa cells cultured on coverslips were fixed with paraformaldehyde, permeabilized with Triton X-100 and stained with various antibodies plus fluorescein—conjugated second antibody. The fluorescence staining was visualized by laser confocal microscopy, collecting images through 9 consecutive focal planes. In this study, ENA anti-Sm was used as a positive control for nuclear staining (Figure 2). In a plane above the cell nucleus, there was little fluorescence. Through the middle sections, the plane of focus cuts through the nucleus and yielded intense staining (Figure 2, column B). One of the middle sections is enlarged in column A (Figure 2), showing diffuse distribution of fluorescence throughout the nucleus, except for the "black holes" (see Figure 2, Chapter 2). Similarly, anti-galectin-3 yielded, through the middle sections, bright nuclear staining with the exception of the "black holes." Thus, the subcellular distribution of galectin-3 and the Sm epitopes of snRNPs are very similar, a conclusion documented in Chapter II. Staining for LDH, serving as a negative control, showed that the enzyme was predominantly cytoplasmic. With the enlarged middle section (Figure 2, column A), the anti-LDH yielded a distinct pattern, showing that the majority of fluorescence in the cytoplasm. The staining for galectin-1 yielded sections that resembled neither those obtained with anti-Sm or anti-galectin-3 nor that seen with anti-LDH. Due to high levels of the lectin 141 Figure 2: The subcellular localization of galectin-1 as revealed by laser scanning confocal fluorescence microscopy. The staining of snRNP Sm polypeptides and galectin-3 and the staining of LDH provided reference pattems for nuclear and cytoplasmic localizations, respectively. For each staining, images (360 x 360 pixels) were collected from 9 consecutive sections focal planes, with the increment of 0.5 pm for each step in the x and y directions. They are displayed serially in column B; column A provides an enlarged view of one middle section (#5). 142 Sm Galectln-S Galectin-1 143 in the cytoplasm, the middle sections yielded generally diffuse fluorescence throughout both the cytoplasm and the nucleus. The areas devoid of fluorescence are not nuclei; rather, they correspond to the "black holes" observed with anti-Sm or anti—galectin-3 staining. 144 DISCUSSION The previous observations on the difference in the ability of Lac-agarose versus anti- Mac 2 to deplete splicing activity from nuclear extracts of HeLa cells suggested: (a) in addition to galectin-3, there must be other Lac-binding proteins in splicing competent extracts; (b) depletion of a single lectin from such an extract cannot remove the splicing activity; and (c) the splicing activity is lost only when all the Lac-binding proteins are adsorbed on the Lac-agarose resin. Coupled with the demonstration that either galectin-1 or galectin-3 can reconstitute the splicing activity in a depleted extract (9), these results suggest that there may be multiple members of the galectin family in the nucleus and that their activities may be redundant. Indeed, the results of this series of experiments have identified at least galectin-1 to be present, in addition to galectin-3, in the bound fraction of the Lac-agarose resin when nuclear extracts are depleted of splicing activity. The localization of galectin-1 to the nucleus was also confirmed by confocal immunofluorescence microscopy. Previous comparisons of the immunofluorescence staining of live cells versus cells fixed with formaldehyde followed by permeabilization indicate that galectin-1 is found predominantly in the intracellular compartment. Within the cells, the staining is mostly cytoplasmic. Observations and interpretations pertaining to the nuclear localization of galectin-1 are somewhat more difficult to establish. First, immunofluorescence and ultrastructural studies have led to explicit statements that anti-galectin-l antibodies failed to label the cell nucleus. In chicken embryo 145 fibroblasts, anti-chicken heart lectin (chicken galectin-1) staining was described to be localized only to the cytoplasm (17). When 17-day-old chicken embryonic keratinized epidermis was stained with gold-labeled anti-galectin-l under electron microscopy, gold particles were found around desmosomes, tonafilament bundles, and the intercellular space, while the cell nucleus was free of the particles (18). Second, in a number of studies originally performed for other objectives (e. g. to show overlap in cells expressing lectins and lactoseries glycoconjugates), the investigators provide no specific conclusion regarding the intracellular distribution of galectin-1. Dorsal root ganglion neurons were subjected to immunofluorescence with anti-galectin-l and with a monoclonal antibody directed against a lactoseries glycoconjugate (19). Although the intracellular distribution of the lectin was not discussed in that particular report, the same investigators have more recently concluded, on the basis of the previously published data, that galectin-1 could be detected in both the nucleus and cytoplasm of the neurons (20). Similarly, reassessment of previously published immunocytochemical studies in non- neuronal cells has suggested the presence of rat galectin-1 in nuclei and cytoplasm (21). Finally, there are studies that specifically show the localization of galectin-1 in the nucleus as well as the cytoplasm. Cryostat sections of tissues subjected to immunofluorescence showed labeling of both nuclei and cytoplasm in anti-chicken galectin- l staining of adult chicken kidney (22) and anti-bovine galectin-1 staining of calf pancreas (23). A monoclonal antibody, designated 36/8, was generated against bovine heart galectin- ] and stained the nuclei and cytoplasm of lymphoblastoid and leukemic cells (24). This monoclonal antibody immunoblotted polypeptides of apparent molecular mass 13, 36, 65, 146 80, and 130 kDa in extracts of lymphoid cells. More recent studies have shown that the 36/8 antibody recognizes the tetrapeptide sequence Trp—Gly-Ala/Ser—Glu/Asp (25) and, therefore, it is not clear whether the nuclear staining component is a galectin or some other polypeptide bearing this epitope sequence. At the ultrastructural level, it has been reported that anti-galectin-l labels the nucleus of epidermal cells of the intermediate layer of a stratified epithelium, the chick embryonic skin (26). On the basis of these previous reports, it seems apparent that the localization of galectin-1 has been somewhat controversial. It was important to establish, therefore, that galectin-1 can indeed be found in the cell nucleus. The criteria used in the present study consisted of parallel labeling studies with antibody reagents directed against known nuclear components (e. g. anti-Sm) and an enzyme generally accepted as a cytosolic marker (e. g. anti-LDH). The staining patterns obtained with anti—galectin-l under confocal immunofluorescence analysis resembled neither anti-Sm nor anti-LDH. Rather, the anti- galectin-l staining represented a composite of the labeling patterns of nuclear and cytoplasmic controls, suggesting that the lectin is found in both compartments of the cell. 10. 11. 12. 13. 14. 147 REFERENCES . Barondes, S. H., Cooper, D. N. W., Gitt, M. A., and Leffler, H. 1994. J. Biol. Chem. 262, 20807-20810. Hirabayashi, J. and Kasai, K. 1993. Glycobiology 3, 297-304. Moutsatsos, I. K., Davis, J. M. and Wang, J. L. 1986. J. Cell Biol. 102, 477-483. Agrwal, N., Wang, J. L. and Voss, P. G. 1989. J. Biol. Chem. _246, 17236-17242. Moutsatsos, I. K., Wade, M., Schindler, M. and Wang, J. L. 1987. Proc. Natl. Acad. Sci. USA 84, 6452-6456. Jia, S. and Wang, J. L. 1988. J. Biol. Chem. 263, 6009-6011. Laing, J. G. and Wang, J. L. 1988. Biochemistry _2_7, 5329-5334. Dagher, S. R, Wang, J. L. and Patterson, R. J. 1995. Proc. Natl. Acad. Sci. USA 22, 1213-1217. Dagher, S. F. 1994. Ph.D. thesis, Michigan State University. Ho, M. K. and Springer, T. A. 1982. J. Immunol. £8, 1221-1228. Cherayil, B. J ., Weiner, S. J ., and Pillai, S. 1989. J. Exp. Med. fl), 1959-1972. Agrwal, N., Sun, Q., Wang, S. Y., and Wang, J. L., 1993. J. Biol. Chem. _2_6_8, 14932- 14939. Metcalf, T. N., Szabo, L. J ., Schubert, K. R., and Wang, J. L. 1984. Protoplasma 1_2§, 91-99. Villanueva, M. A., Ho, S. -C. and Wang, J. L. 1990. Arch. Biochem. Biophys. _2_71, 35- 41. 15. 16. 17. 18. 19. 20.: 21. 22. 23. 24. 25. 26. 148 Dignam, J .D., Lebovitz, RM. and Roeder, R.G. 1981. Nucleic Acids Res. 11, 1475- 1489. Wade, M. H., Defeijter, A. W., Frame, M. K., and Schindler, M. 1993. Bioanlyt. Instrumen. 31, 117-141. Barak-Briles, E., Gregory, W., Fletcher, P. and Komfeld, S. 1979. J. Cell Biol. 8_1, 528-537. Hirano, H., Akimoto, Y., Kawakami, H., Oda, Y. and Kasai, K. 1988. in Proceedings of the Kagoshima International Symposium of Glycoconjugates in Medicine, Professional Postgraduate Service International, Tokyo, pp. 8-13. Regan, L. J ., Dodd, J., Barondes, S. H., and Jessell, T. M. 1986. Proc. Natl. Acad. Sci. USA 83, 2248-2252. Hynes, M. A., Gitt, M., Barondes, S. H., Jessell, T. M. and Buck, L. B. 1990. J. Neurosci._1_Q, 1004-1013. Barondes, S. H. 1988. Trends Biochem. Sci. l_3, 480-482. Beyer, E. C. and Barondes, S. H. 1980. J. Supramol. Struct. 13, 219-227. Childs, R. A. and Feizi, T. 1980. Cell Biol. Int. Rep. 4, 775. Carding, S. R., Thorpe, S. J ., Thorpe, R. and Feizi, T. 1985. Biochem. Biophys. Res. Commun. l_27, 680-686. Abbott, W. M., Mellor, A., Edwards, Y. and Feizi, T. 1989. Biochem. J. _2_52, 283-290. Akimoto, Y., Kawakami, H., Oda, Y., Obinata, A., Endo, H., Kasai, K., and Hirano, H. 1992. Exp. Cell Res. 1%, 297-304. CHAPTER V Concluding Statement Removal of intron and ligation of exons is one of the most important steps for pre- mRN A processing. Many catalytic elements have been elucidated for their involvement in splicing machinery. Previous studies have shown that galectin-3 is a required factor for spliceosome formation and splicing activity using a cell-free assay for pre-mRNA splicing. However, the subnuclear localization and the RNP components which galectin-3 is involved in pre-mRN A splicing remain unclear. To address these questions, I have developed two levels of study to demonstrate that galectin-3 is associated with components of the splicing machinery, specifically, the Sm epitopes found on polypeptides of the snRNPs. At the single cell level, the iMunofluorescence patterns of galectin-3 were shown to be similar to that of the snRNP Sm polypeptides during the course of cell cycle and under a variety of conditions. Double labeling experiments also demonstrated that galectin-3 colocalized with Sm antigens in the cell nucleus. At the biochemical level of protein-protein interaction, I have shown that a monoclonal antibody anti-Mac 2, which is specific for galectin-3, could coimmunoprecipitate a fraction of Sm B polypeptides from nuclear extracts of HeLa cells and mouse 3T3 fibroblasts. Nuclear extracts depleted of galectin-3 by prior adsorption on a lactose affinity resin failed to yield Sm B in the anti-Mac 2 precipitate. These results demonstrate that the coprecipitation of Sm B with galectin-3 by anti-Mac 2 was dependent 149 150 demonstrate that the coprecipitation of Sm B with galectin-3 by anti-Mac 2 was dependent on the presence of the lectin. The coprecipitation was not affected by the addition of lactose; nor was it perturbed by prior treatment of the nuclear extract with ribonuclease. Taken together, these results suggest that at least a fraction of the Sm B polypeptides in the splicing extract can interact with galectin-3 through protein-protein interactions. Identification of the RNP component with which galectin-3 is associated should be the next level of study to address the function of galectin-3 in the splicing machinery. I have also analyzed the composition and identities of polypeptides bound to the affinity resin lactose-agarose under conditions used to deplete the nuclear extract of splicing activity. In addition to galectin-3, the bound fraction of the lactose-agarose also contains galectin-1, whose presence in the nucleus of HeLa cells was confirmed by confocal fluorescence microscopy. These findings provide an explanation for previous observations that while lactose-agarose can deplete nuclear extracts of splicing activity, anti-Mac 2 precipitation failed to yield the same effect. Together with the observation that galectin-1, as well as galectin-3, can alone reconstitute splicing activity in a lactose-agarose depleted extract, these results suggest that the activities of galectin-1 and galectin-3 are redundant in the cell nucleus. MICHIGAN 511an UNIV. LIBRARIES llllllllll lllllll 1111111111 1111111 11111111111111 31293014172252