_. afiwm? "Eng 5”“ J”. .54. 4. I»! « .4: in... L3 xunuanrnmnwflflrs. . . id‘sl. .- fc?{r Ihufmg V . . ulnflr‘w‘fi:ltafl. , y . ‘ \l, A 1v [A1, . . ‘ . .....xhuwh... V humufitn .. ‘ . . . . «.1 aduFCJ‘ia v: 5%“: 0..., in :«I 51., I . a: 4 .53.}. :5: t”. Lama? than" r... .15... LA. I. an... .. a a. :nae. : 2.. .. H.351; , 15.13:,"- .32 if... a i 3.??? 3. ,7.-. .5: 713.1: I! I... .61.. :1 ‘ .. . “Pb:- a ... t. .2; L. 13,133.: I 3 . 1.1.12}? :33 5...... LI- l:¥ll$.....5‘ )1?! l? 1 I}... . 0.. :5: .1! 1 3E. :._ ..A I» no: u .l ~4m ‘ .. , . r . .fi. .. HF team"; ~35... Enxxfikr. E agghczfiwaaé E {FR 6 THESIS K7 V 2550 LIBRARY Michigan State University This is to certify that the dissertation entitled CLUM/vé. AND CHARACTZ>RIZATIDM 0F CANINE TNTRJN§lC FACTDR- (Loam/Maw RECEPTOR/cueiLw presented by ’DANBt/V XH has been accepted towards fulfillment of the requirements for ‘DDCTDR degreein PHILOSOPHY SL—C/W V Major p‘rofessg {1- (#00 Date MS U is an Affirmative Action/Equal Opportunity Institution 0-12771 PLACE IN RETURN BOX to remove this checkout from your record. TO AVOID FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE DATE DUE 6/01 cJCIRC/DateDuepes-p. 15 CLONING AND CHARACTERIZATION OF CANINE INTRINSIC FACTOR- COBALAMIN RECEPTOR I CUBILIN By Danbin Xu A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Microbiology 2000 ABSTRACT CLONING AND CHARACTERIZATION OF CANINE INTRINSIC FACTOR- COBALAMIN RECEPTOR I CUBILIN By DANBIN XU Cubilin is a high molecular weight multiligand receptor that mediates intestinal absorption of intrinsic factor-cobalamin and selective protein reabsorption in renal tubules. The genetic basis of selective intestinal cobalamin malabsorption with proteinuria was investigated in a canine model closely resembling human lmerslund-Grasbeck syndrome caused by cubilin mutations. Canine cubilin CDNA of 11,282 bp was cloned and sequenced, including 73 bp of 5’ and 349 bp of 3’ untranslated sequence. The open reading frame of 10,860 bp corresponds to a deduced amino acid sequence of 3,620 residues and demonstrates an overall amino acid identity of 83% with human and 70% with rat cubilin. Further characterization of tissue-specific cubilin expression and post- translational modification with emphasis on the gastrointestinal tract was carried out. Intrinsic factor-cobalamin binding activity, cubilin immunoreactivity, and cubilin mRNA levels were determined in multiple segments of canine gastrointestinal mucosa and other tissues. The parallel aspects of cubilin expression suggested that the major determinant of regional cubilin expression in the gastrointestinal tract was modulation of cubilin mRNA. Cell fractionation indicated that ileal cubilin is not strongly membrane-associated. An ~185 kDa brush-border specific and two >400 kDa precursor forms of cubilin were identified in ileum, but only an ~460 kDa form in renal cortex. Asparagine-linked oligosaccharide modifications characterized by differential glycosidase digestion of affinity purified cubilin from ileal mucosa and renal cortex. Neal and renal intracellular cubilin comigrated on SOS-PAGE at ~4OO kDa after oligosaccharide removal, suggesting that the size differences were caused by different glycosylation. lntragenic markers were identified within introns for both cubilin and megalin genes in the canine family and used to test the hypothesis of genetic linkage of the disease to cubilin or megalin loci. Megalin, a transmembrane receptor interacting with cubilin in vitro and colocalizing with cubilin in vivo, is speculated to anchor cubilin onto apical membranes of ileal enterocytes. Linkage of either gene was rejected, indicating that the canine disorder resembling Imerslund-Grasbeck syndrome is caused by defect of a gene product other than cubilin or megalin. These results imply that there may be locus heterogeneity among human kindreds with selective intestinal cobalamin malabsorption and proteinuria and that normal brush-border expression of cubilin requires the activity of an accessory protein other than megalin. This dissertation is dedicated to: Qiang Tian, my husband and best friend, for his deep love, care and support through all my scientific growth. John Fyfe, my mentor and friend, for his patience, encouragement and understanding during my graduate studies. iv ACKNOWLEDGMENTS I am forever grateful to the large number of people who gave me advice, assistance and encouragement during the completion of my Ph.D. I feel fortunate to have had the support and friendship of so many wonderful people. I am especially grateful to the members of my graduate committee, Dr. Jerry Dodgson, Dr. Ronald Patterson, Dr. Richard Schwartz and Dr. Laura McCabe, for their guidance, instruction and encouragement throughout this research work, for the valuable time and comments on this dissertation. I want to thank Rebeccah Kurzhals and Dr. Mary Lassaline for their friendship and the help for taking care of the dogs. I am also thankful to Dr. Karen Friderici and people in her lab for the use of laboratory equipment, supplies, and technical knowledge. I also owe thanks to several members of the Department of Microbiology, Dr. Walt Esselman, Angie Zell, and Cheryl Akers for their help during my graduate studies. I also thank my fellow students for their friendship, and support during my years at Michigan State University. I like to thank Dr. Pierre Verroust, Dr. Renata Kozyraki in France for providing the rat cubilin CDNA, and Dr. Paula Henthorn, Dr. Jianlong Liu in University of Pennsylvania for providing the technical support. Special thanks also go to my wonderful family, my parents and my sister for their unconditional support and encouragement throughout all my endeavors in life. A very special thank due to my lovely daughter, Elaine, for the cheerful moment she gave me when I had hard time for the research. TABLE OF CONTENTS LIST OF FIGURES .......................................................................................... viii CHAPTER 1 LITERATURE REVIEW ........................................................................................ 1 Overview of normal cobalamin absorption ................................................... 1 Cobalamin malabsorption and Imerslund-Gréisbeck syndrome .................. 7 Human lmerslund-Grasbeck syndrome .......................................................... 7 Canine model with selective cobalamin malabsorption ................................... 9 Characterization of intrinsic factor-cobalamin receptor (IFCR)Icubilin ....11 General characteristics of IFCR I cubilin ....................................................... 11 Molecular cloning of human and rat cubilin ................................................... 14 Functional domains of cubilin ......................... - ............................................... 1 9 Regulation of cubilin expression in small intestine .................................... 21 Protein folding in the secretory pathway ..................................................... 24 General outline for this thesis ...................................................................... 28 References ..................................................................................................... 30 CHAPTER 2 CLONING OF CANINE CUBILIN ....................................................................... 43 Introduction .................................................................................................... 44 Materials and Methods .................................................................................. 46 Results ............................................................................................................ 50 Discussion ...................................................................................................... 55 References ..................................................................................................... 59 CHAPTER 3 CUBILIN EXPRESSION AND POST-TRANSLATIONAL MODIFICATIONS IN THE CANINE GASTROINTESTINAL TRACT .................................................... 61 Introduction .................................................................................................... 62 Materials and Methods .................................................................................. 65 Results ............................................................................................................ 74 Discussion ...................................................................................................... 94 vi References ................................................................................................... 103 CHAPTER 4 LINKAGE ANALYSIS OF CANINE SELECTIVE COBALAMIN MALABSORPTION WITH CUBN AND MEGALIN LOCI ................................. 107 Introduction .................................................................................................. 108 Materials and Methods ................................................................................ 1 1 1 Results .......................................................................................................... 1 16 Discussion .................................................................................................... 123 References ................................................................................................... 127 Future Direction .............................................................................................. 130 vii LIST OF FIGURES Figure 1.1 Canine intrinsic factor-mediated cobalamin absorption .............. 2 Figure 1.2 Predicted protein structure of cubilin ........................................... 15 Figure 1.3 Schematic representation of the interaction between cubilin and megalin ...................................................... 19 Figure 2.1 CDNA cloning of canine kidney cubilin ........................................ 51 Figure 2.2 Primary sequence of canine cubilin as deduced from CDNA clones ................................................... 52 Figure 3.1 Tissue specificity of cubilin mRNA expression ............................ 75 Figure 3.2 Codistribution of intestinal cubilin function, immunoreactive proteins, and mRNA ......................................... 76 Figure 3.3 Codistribution of quantitated intestinal cubilin ligand-binding activity and mRNA .................................... 80 Figure 3.4 lmmunoreactive and lF-Cbl binding cubilin species in fractionated canine ileal mucosa and renal cortex .......................................................................... 83 Figure 3.5 Western blot of canine cubilin in reducing and non-reducing conditions ...................................................... 85 Figure 3.6 Distribution of immunoreactive cubilin forms in different centrifugation fractions of dog ileal homogenate ........................................................................ 87 Figure 3.7 Oligosaccharide analysis of affinity purified ileal and renal cubilin .................................................................. 88 Figure 3.8 Northern blot analysis of canine cubilin and CRP-ductin along the crypt-villus axis in dog ileum .................... 90 Figure 3.9 Alkaline phosphatase activity along the crypt- villus axis in dog ileum ................................................................ 91 Figure 4.1 Independent segregation of canine I-GS and CUBN loci ............ 117 Figure 4.2 lntragenic cubilin marker is located in a unique region of the canine genome .......................................... 118 viii Figure 4.3 Independent segregation of canine l-GS and megalin loci ......... 120 ix CHAPTER 1 LITERATURE REVIEW Overview of normal cobalamin absorption Cobalamin (Cbl) is a complex organometallic substance consisting of a corrin ring, a central cobalt atom, and various axial ligands. The basic structure, known as vitamin B12, is synthesized exclusively by microorganisms. ln non-ruminant animals, bacterial synthesis takes place only in the large bowel and the cecum, and from these sites absorption cannot take place. Therefore, absorption of vitamin B12 is entirely from dietary sources (Booth and Mollin, 1959), including fecal contamination or coprophagy. Cbl is an essential micronutrient required for the health and well being of all higher animals, including human. All of these animals are capable of converting the vitamin into the two required coenzyme forms, adenosyl-cobalamin (AdoCbl) and methylcobalamin (Merl), which are essential cofactors for methylmalonyl-CoA mutase and methionine synthetase, respectively, in intermediate metabolism. Cobalamin absorption, which is a complex process comprised of sequential protein-binding events, begins with a digestive phase (Figure 1.1). The vitamin is bound predominantly by haptocorrin (HC, R protein) in gastric juice and becomes bound to intrinsic factor (IF) after the HC moiety is partially degraded by pancreatic proteases in the duodenum (Allen et al., 1978; Carmel et al., 1983). IF is a Cbl-specific-binding protein classically synthesized in man by the gastric parietal cells of the fundus and body of the stomach (Christensen et al., 1973; Levine et al., 1980). IF is synthesized by various cells and glands Target Tissues Cbl II" IFCR TC-II ac Stomach D b7 IF-Cbl Complex Y V O Bi] Degraded IF . D 3 Pancreas Protease: Q9 Degraded ac D ’ a ’ 1' Undefined Hechanim Blood Side Figure 1.1 Canine intrinsic factor-mediated gastrointestinal cobalamin absorption. Provided by Dr. John Fyfe. ' derived from foregut depending on the different species (Gordon, 1989; Festen, 1991). IF is produced by the gastric mucosa of humans, pigs, and rats but is produced mainly in pancreatic duct cells in dogs (Batt and Horadagoda, 1989; Simpson et al., 1989; Vaillant et al., 1990; Simpson et al., 1993) and pancreas in cats (Fyfe, 1993). IF synthesized by different species showed different cross- species immunoreactivity (Hooper et al., 1973). Functional IF secretion and IF mRNA levels are developmentally regulated (Ramaswamy et al., 1989; Dieckgraete et al., 1988). IF secretion is also tissue specifically regulated, being activated by pentagastrin in the human stomach and by cholecystokinin in the canine pancreas after eating (Simpson et al., 1993), but the correlation between IF secretion and the expression of other Cbl-binding proteins is still unknown. The lF-Cbl complex is highly resistant to proteolysis. This enables it to pass intact through the small bowel. Physiological cobalamin absorption takes place in the distal small intestine (Booth and Mollin, 1959), which has specific receptors for the IF-Cbl complex on the microvillous membranes of mucosal cells (Donaldson et al 1967; Levine et al., 1982). Attachment of the IF-Cbl complex to the receptor requires the presence of divalent cations (notably calcium) (Mackenzie et al., 1972) and a pH between 6 and 9 (Cooper and Castle, 1960; Herbert and Castle, 1961) but is independent of temperature and energy (Donaldson et al., 1967). Intrinsic factor-cobalamin receptor (IFCR), a membrane protein of the ileal brush border, mediates intestinal uptake and initiates transcytosis of dietary and biliary Cbl across the mucosal barrier. The number of receptors in the intestine seems to be very low. The greatest density of ileal receptor sites is exhibited in dogs and pigs, being approximately 0.3 x1012 to 4.9 x 1012 binding sites/ gram mucosa (Hooper et al., 1973). The restricted absorption of cobalamin is partly explained by the small number of receptors. IFCR is specific for IF-Cbl complex while free cobalamin, free IF and Cbl attached to other Cbl binding proteins cannot bind the receptor (Hooper et al., 1973; Katz and cooper, 1974). There is an enterohepatic cycle of Cbl absorption. The daily amount of Cbl excreted in bile constitutes approximately 0.1 to 0.19% of the total amount stored in the human body (Nicolas JP and Guéant JL, 1995). Pancreatic enzymes are responsible for the degradation of biliary R protein leading to the transfer of the released Cbl to biologically active IF and eventually to Cbl absorption in the terminal ileum (El Kholty et al., 1991). Following the adsorption of the IF-Cbl complex to the ileal receptor there is a delay of 3 to 4 hours before the vitamin appears in the blood (Doscherholmen et al., 1959; Fyfe et al., 1991a). The processes involved in the translocation of cobalamin from the ileal receptor to the blood are still largely unknown, though some knowledge has accumulated. The receptor mediated endocytosis of IF-Cbl complex occurred within 30 min after the complex attached to its ileal surface receptor (Kapadia et al., 1983). In vivo and in vitro studies with 57Co-Iabeled cyanocobalamin and [35$]methionine-Iabeled IF showed that following endocytosis, the IF-cobalamin complex was released from the receptor and transferred to the lysosomal or prelysosomal acidic vesicles, where IF was degraded with a half-life of 4 hours (Kapadia et al., 1983; Ramanujam et al., 1991a, Guéant et al., 1992; Dan at al., 1994; Bim et al.,1997). Chloroquine or cycloheximide treatment reduced the transport of Cbl suggesting release of Cbl from IF required acid pH, that there was a series of compartments involved in the transport of Cbl, and that new protein synthesis was required (Robertson JA., 1985a; Robertson JA, 1985b; Ramaswamy et al., 1989) The receptor of lF-Cbl complex is rapidly recycled onto the brush-border membrane after endocytosis (Dan and Cutler, 1994; Le Panse et al., 1997), and the liberated Cbl exported from Iysosomes via a specific transporter ( that malfunctions in Cbl F) to transcobalamin ll (TC II). The location for this transport is still an open question. Some evidence suggested that Cbl released from IF and transfered to TC II within the rat enterocytes (Robertson and Gallagher, 1985b; Ramaswamy et al., 1989; Ramanujam et al., 1991a). But other researchers questioned if the crude homogenate used for TC lI-[57Co]CbI immunoprecipitation can provide sufficient discrimination between the tissue components of the ileum (Quadros et al., 1999). TC II is a specific Cbl transport protein which has been detected in many cell types in culture (Hall et al., 1985) and in human and rat tissues (Li et al., 1994). In dogs, all the plasma Cbl is bound to TC-Il (Rappazzo and Hall, 1972). In contrast, in human, only newly absorbed Cbl is bound to TC-ll, and most circulating Cbl is bound to TC-l which is haptocorrin (HC) (Gullberg R., 1972). This distinction is important since, in dogs, TC-II deficiency would be expected to have extremely low serum Cbl concentration, but in human TC-II deficiency, serum Cbl concentrations are similar to normal due to Cbl binding to HC (Fenton and Rosenberg, 1989). The canine TC II has been studied and showed similar molecular size , cobalamin binding activity, and immunologial reactivity as in human (Rappazzo and Hall, 1972). TC Il-mediated uptake of plasma Cbl has been shown in a number of cells (Hall 1975; Retief et al., 1966; YoungdahI-Turner et al., 1978), and Cbl is transported into cells via a plasma membrane TC II-Cbl receptor (TC ll-R) expressed in many tissues (Bose et al., 1995a; Bose et al., 1995b). After endocytosis, the TC Il-Cbl complex dissociates in endosomes or Iysosomes, and free Cbl is released (Idriss et al., 1991). The free Cbl then enters the cytoplasm where it is reduced and converted to methyl-Cbl, or in mitochondria, where it is reduced and converted to adenosyl-Cbl. Methyl-Cbl and adenosyI-Cbl are used as cofactors for methionine synthase and methylmalonyl CoA mutase, respectively (Millman et al., 1977, Kolhouse and Allen, 1977). In addition to TC II- R, whose role in plasma transport of Cbl is well established, another protein known as megalin has been shown to bind to TC Il-Cbl in vitro and mediate its endocytosis in BN/MSV cells (derived from yolk sac carcinoma) and perfused renal proximal tubules (Moestrup et al., 1996). The physiological significance of megalin-mediated TC II-Cbl uptake system in the plasma transport of Cbl is not certain. Due to the high expression in the apical membrane of kidney proximal tubules, renal megalin may function in the tubular reabsorption of TC II-Cbl, thus preventing Cbl loss in the urine. Cobalamin malabsorption and lmerslund-Grfisbeck syndrome Due to the widespread distribution of cobalamin in animal products (Sullivan LW, 1970; Smith A, 1962), Cbl deficiency usually occurs secondary to inherited or acquired defects of gastrointestinal Cbl absorption rather than dietary insufficiency. Inherited disorders of vitamin B12 include those in which the vitamin is not utilized by target cells, and those which are defective in absorption of the vitamin from the gut and transport to the appropriate tissues. The former includes a defect in the release of free vitamin B12 from Iysosomes (CbIF) (LaFramboise et al., 1992) and defects in the formation of one or both vitamin B12 cofactors (CbIA- E). Defects of absorption and transport include intrinsic factor abnormalities (Spurling et al., 1964; Katz et al., 1974b; Yang et al., 1985), deficiencies of TC II (Hakami et al., 1971) Cbl F, and selective intestinal Cbl malabsorption. Human lmerslund-Grfisbeck syndrome Selective intestinal Cbl malabsorption is also called Imerslund-Grasbeck syndrome (I-GS) which was independently described by Imerslund and Grasbeck in 1960 (Grasbeck et al., 1960, Imerslund 0., 1960). lmerslund’s ten patients were from six different families in south-east Nonlvay, while Grasbeck et al. reported two cases from Finland. The patients examined showed Cbl-responsive anemia and most of them also had mild proteinuria. Following thorough investigation, all known causes of Cbl deficiency were eliminated, including pathological intestinal flora, inactive intrinsic factor, and deficiency of transcobalamin I or II. Both sexes were affected, and the inheritance was deemed recessive. The similarity of the two sets of cases suggested that they represented the same disease. Since then, totally 38 cases in Finland and 15 cases in Norway have been identified with selective Cbl malabsorption (Aminoff et al., 1995). More cases have also been reported in other countries. By 1969, 20 cases with l-GS had been diagnosed in Israel, and proteinuria showed predominantly albumin (Ben-Bassat et al., 1969). Other scattered cases were also described from the united state (Colle et al., 1961; Spurling et al., 1964), Denmark (Sievens CJ, 1964), Australia (Marsden et al., 1979), Turkey (Yetgin et al., 1983; Altay et al., 1995; 1999; Celep et al., 1996), and Saudi Arabia (Abdelaal and Ahmed, 1991). More recently a single case has also been reported in an Africa (Stones and Ferreira, 1999). By 1999, there were over 250 cases reported in the literature. The syndrome occurs worldwide, but its prevalence is higher in several Middle Eastern countries and Norway, and highest in Finland. The onset of signs of this disease is between 0 to 5 years of age, which mainly corresponds to the expected period of depletion of the child’s vitamin B12 which was stored prenatally by transfer from the maternal blood. The clinical features of this syndrome include failure to thrive, infections, megaloblastic anemia, neuropathy and mild general malabsorption. Mild proteinuria is common. All the clinical features except Cbl malabsorption and proteinuria can be eliminated by parenteral, but not oral, Cbl administration (Broch et al., 1984). Linkage analysis of 38 patients diagnosed in Norway and Finland localized the unknown autosomal recessive megaloblastic anemia (MGA 1) gene to the short arm of chromosome 10 between markers D108548 and D1OS466 (Aminoff et al., 1995). Due to the clinical heterogeneity of this syndrome among the affected families and complexity of the process of enterocyte transcytosis of Cbl, different putative molecular natures of Imerslund-Grasbeck syndrome have been reported. One study showed lack of pathology by electron microscopy of ileal biopsies and normal lF-Cbl binding in mucosal homogenates and suggested the cause of this disease has a defect in intracellular trafficking of Cbl rather than a deficient synthesis of the receptor (Mackenzie et al., 1972). Another study in a different family showed defective uptake of Cbl by ileal mucosa in vivo (Burman et al., 1985). The difference between these two studies may be due to the different genetic defects, but is also possibly due to the same genetic defect but different study design. Canine model with selective cobalamin malabsorption A similar recessively inherited syndrome in giant schnauzer dogs has been reported, in which affected puppies exhibited inappetance and failure to thrive beginning between eight and twelve weeks of age. (Fyfe et al., 1989). Clinical laboratory tests demonstrated megaloblastic anemia, low serum Cbl concentrations, and mild proteinuria. Parenteral, but not oral, administration of cyanocobalamin corrected all clinical and laboratory abnormalities except proteinuria and Cbl malabsortption. Gastrointestinal cobalamin malabsorption was demonstrated by oral radiolabeled cobalamin administration, and biochemical characterization of IF and TC II were normal (Fyfe et al., 1991a). Expression of IFCR was investigated in ileum and renal cortex of affected dogs by immunoelectron microscopy and cell fractionation, and the results demonstrated that there was no expression of IFCR in the apical brush-border membrane of either tissue (Fyfe et al., 1991b). Receptor-ligand binding assay showed IFCR activity in total homogenates was 3-4 fold higher than normal in ileal mucosal and one-tenth of normal in kidney of affected dogs. These data suggested that inefficiently expressed receptors accumulate in ileum but were rapidly degraded in kidney of affected dogs (Fyfe et al., 1991 b). Brush-border expression of IFCR is critical for intestinal IF-Cbl absorption. The defective functional expression of IFCR onto the apical membrane raised the consideration that IFCR may be a candidate gene for MGA 1 in this family of dogs. The most common reason for disfuntional membrane expression of a membrane protein is mutation of the coding sequence, which results in abnormal protein folding (discussed later). Based on this phenomenon, we hypothesized that mutation of IFCR was the cause of selective intestinal Cbl malabsorption in this family of dogs. The similarity of the clinical and laboratory features between human patients and the dog family with selective intestinal Cbl malabsorption suggested that this would be a good animal model for study of this syndrome. Due to the easy treatment of the disorder in humans, experimental samples are extremely hard to obtain from patients, greatly limiting investigation of the molecular mechanism of this disease. Biochemical study of IFCR in the affected dogs first demonstrated the candidate gene for MGA 1. Further study with this model will enhance understanding of this syndrome and elucidate mechanisms of the cellular processing pathway of newly synthesized proteins. Characterization of intrinsic factor-cobalamin receptor (IFCR)Icubilin General characteristics of IFCR I cubilin Clearly IFCR plays a key role in gastrointestinal Cbl absorption. Research has been done during the past over 25 years to further understand the function and molecular nature of this receptor. Solubilization of IFCR from distal small intestine has been carried out in guinea pig and pig (Katz et al., 1974a; Cotter and Rothenberg et al., 1976). Solubilized receptor had similar IF-Cbl binding characteristics to membrane-bound receptor, suggesting the functional domain of this receptor resided on the Iumenal side of the enterocyte membrane. Canine ileal IFCR purified by affinity chromatography revealed a 180 kDa protein on non-reducing SDS-PAGE, and 2-mercaptoethanol treatment produced two bands with apparent molecular weights of 59 and 42 kDa (Seetharam et al., 1981). Papain digestion prior to purification resolved IFCR into smaller subunits of the receptor which maintained IF-Cbl binding activity, both for dogs (Seetharam et al., 1982) and pigs (Kouvonen I, 1980; Kouvonen and Grasbeck, 1980). Further cell fractionation of canine ileal enterocytes revealed an ~185 kDa lF-Cbl binding protein on brush-border membrane (Fyfe et al., 1991b). However, functional IFCR in the Caco-2 cell line, derived from a human colon carcinoma, was reported as a 230 kDa protein (Ramanujam KS, 1991a) which does not dissociate into subunits upon disulfide bond reduction. This suggested that, instead of having a subunit structure, IFCR undergoes proteolytic cleavage once exposed to the ileal lumen with the resulting fragments being held together by disulfide bonds. Besides the distal part of the small intestine, high levels of receptor for IF- Cbl were detected in human, canine, and rat kidney. The ratio of specific activity for kidney relative to intestine in these species was 116, 20 and 797, respectively (Seetharam et al., 1988). In vivo (Seetharam et al., 1992) and in vitro (Ramanujam et al., 1991b, 1992) functional studies of renal IF-Cbl receptor showed the same binding activity and immunoreactivity as the ileal receptor. Both activities were detected in the apical membrane of proximal tubule cells but not in distal tubule cells of the kidney. The size determined from SDS-PAGE for the renal receptor was reported as 230 kDa and was not reduced into subunits by reductive alkylation (Seetharam et al., 1988; Ramanujam et al., 1993a; Fyfe et al., 1991b). Quantitative amino acid analysis of the renal receptor gave a value of 457,310 g of amino acid / mol of IF-Cbl binding activity but was thought to represent a dimer (Seetharam et al., 1988). All the ileal and renal IFCR data together suggested that IFCR undergoes proteolytic cleavage once exposed to the ileal lumen rather than having a subuit structure. This hypothesis is based on the assumption that ileal and renal IFCR are the same gene products. lF is not thought to enter the circulation system during ileal endocytosis. The presence of IFCR in the kidney suggested that renal IFCR had functions other than renal lF-Cbl reabsorption. 12 In the meantime, the study of an independent group described a 280-kD protein (gp280), defined by monoclonal antibodies raised against rat renal brush- border, which was found in the intennicrovillar domain of the proximal tubule cells of kidney and rat yolk sac epithelium (Sahali et al., 1988). Antibodies to gp280 induced fetal malformations via trapping of the target antigen in the early endocytic compartment thus preventing its normal function in lysosomal transfer (Le Panse et al., 1994; 1995). The function of gp280 was thought to be a receptor, but its ligand(s) was not known until 1997. Rat renal gp280, like the IFCR, binds to the IF-Cbl complex with an association constant of 0.3 x 109 M'1 and mediates its internalization (Seetharam et al., 1997). In addition, antibodies against purified gp280 and IFCR can inhibit the binding of lF-[57Co]Cbl complex to intestinal, renal, and yolk sac apical membranes and revealed a single identically sized protein on immunoblotting of renal membranes, suggesting functional and immunological identity of the two proteins. The similar mobility on SDS-PAGE and restricted tissue distribution of gp280 and IFCR suggested the possibility that IFCR and gp280 were similar or identical proteins. IFCR/gp280 was then considered an over 200 kDa protein facilitating the intestinal endocytosis of IF-Cbl and renal reabsorption of some unidentified proteins. In late 1997, using receptor-associated protein (RAP) affinity chromatography, an lF-Cbl binding renal epithelial protein of ~460 kDa was co- purified with the TC Il-Cbl binding 600 kDa receptor, megalin (Birn et al., 1997). The IF-Cbl binding activity was further confirmed by IF-Cbl affinity chromatography, which eluted the same ~460 kDa protein from rabbit and human 13 kidney homogenate. In vitro binding analysis demonstrated a calcium-dependent high affinity binding of lF-Cbl to a site distinct from the RAP binding site. Electrophoretic mobility and immunoreactivity was compared between rat gp280 and rabbit ~460 kDa IF-Cbl-binding protein, and the two proteins showed identical SDS-PAGE migration and immunoreactivity. Microscopic autoradiography revealed the ~460 kDa protein in both kidney and terminal ileum and in vivo microinjection demonstrated the facilitation of the endocytosis of IF- Cbl by this membrane protein. Molecular cloning of human and rat cubilin The molecular characterization of this ~460 kDa protein (IFCR/gp280) was not solved until its recent cDNA cloning from rat and human (Moestrup et al., 1998; Kozyraki et al., 1998). The cDNA clone corresponds to a single transcript of 11.6 kb in intestine, kidney and yolk sac tissues in rat, where IFCR/gp280 is expressed. Using fluorescence in situ hybridization, radiation hybrid mapping and screening of YAC clones, the human cDNA clone was mapped on the short arm of chromosome 10 (10p12-p14) (Kozyraki et al., 1998), which is in the same region as human MGA 1. These results suggested that this ~460 kDa protein was the candidate disease-causing gene product for selective intestinal Cbl malabsorption in some patients. All these data showed that the newly purified protein had the same ligand binding activity, tissue distribution, and immunoreactivity, but had different molecular weight compare to IFCR/gp280. Since two very well characterized markers bigger than 460 kDa were used in the 14 most recent study (Birn et al., 1997), the previous over 200 kDa molecular weight for IFCR/gp280 was possibly misjudged by relying on migration on SDS-PAGE with 200 kDa as the highest molecular weight marker. The assembled cDNA revealed an uninterrupted open reading frame of 10.8 kb and a deduced 3621-3623 amino acid protein for human and rat, respectively. The overall homology between the encoded proteins of these two species is 69%. The predicted domain organization of this protein contains an N- terrninal stretch of approximately 110 amino acids with no apparent homology to known proteins, a cluster of eight EGF (epithelial growth factor) type B repeats and 27 contiguous CUB domains (Moestrup et al., 1998; Kozyraki et al., 1998)(Fig 1.2). Due to the unique tandem arrangement of CUB domains in the protein sequence, the ~460 kDa protein (IFCR/gp280) has been designated as cubilin, which will be used for the remainder of this thesis. 8 EGF Repeats 27 CUB Domains WI Figure 1.2 Predicted protein structure of cubilin More evidence suggests that the protein expressed in intestine and kidney are the same gene product. An early study (Robin et al., 1974) reported that a patient with IG-S had an inherent renal proximal tubular defect in amino acid reabsorption which was not Cbl deficiency related. The patients with selective Cbl malabsorption with proteinuria have reduced urinary IFCR activity compared 15 to the normal control (Dugue et al., 1998). Further genetic studies with 17 Finnish families with selective intestinal Cbl malabsorption identified two independent disease-specific mutations in the cubilin gene (CUBN) (Aminoff et al., 1999) . One was a missense mutation (FM1) apparently in the IF-Cbl binding site (Kristinasen et al., 1999). The other mutation was a C to G mutation activating a cryptic intronic splice site and introducting multiple stop codons in the mutant transcript (FM2). These mutations indicated that defects of cubilin in the intestine caused Cbl malabsorption. The Finnish patient with the FM2 mutation also secreted dramatic amounts of urinary alipoprotein A-l (apoA-l) which was not detected in the normal control or FM1 patient (Kozyraki et al.,1999). ApoA-l was also detected in urine of dogs (five of six) with selective intestinal Cbl malabsorption, caused by disfunctional expression of IFCR on both intestinal and renal epithelial cell membranes, but was not detected in normal control dogs. These results elucidated that a null mutation in CUBN (FM2) affects cubilin functions in both intestine and kidney and elegantly supports the hypothesis that these two proteins are the same gene product. The patient with apoA-1 proteinuria indicated that besides lF-Cbl complex, apoA-1 is another ligand for cubilin which can be reabsorbed in the kidney. In vitro studies, such as cubilin affinity chromatography and surface plasmon resonance analysis, further demonstrated the high-affinity binding of apoA-I and high-density Iipoprotein (HDL) to kidney cubilin (Kozyrak et al., 1999; Hammad et al., 1999; 2000). Cubilin-expressing yolk sac cells showed efficient 125l-HDL endocytosis that could be inhibited by antibodies against apoA-l and cubilin. l6 Other ligands have also been defined for renal cubilin recently, such as myeloma light chains (Batuman et al., 1998) and albumin (Birn et al., in press). Both human and rat cubilin open reading frame begin with a 24-20 amino acid signal peptide which facilitates binding of the translating ribosome to bind the endoplasmic reticulum (ER) and starts the membrane protein biosynthetic pathway. An Arg-X-Arg/Lys-Arg consensus sequence can be found following the signal peptide in both sequences, which is a site cleaved by the trans-Golgi proteinase furin (Kozyraki et al., 1998; Moestrup et al., 1998). Cubilin N-terminal sequence starts just after furin site. This indicates that the receptor is synthesized as a precursor undergoing further proteolytic processing in the trans- Golgi. Two of the EGF repeats contain the discontinuous consensus sequence for Cay-binding, which may be required for a particular conformation for ligand binding. The CUB domains, named as a structural feature common to complement subcomponents C1rlC1s, the Xenopus patterning protein, Uvs.2, and Bone morphogenic protein-1, have ~110 residues with two disulfide bridges, and bind a diverse array of protein, carbohydrate, and phospholipid ligands (Bork and Beckmann, 1993). CUB domains are common to an increasing number of developmentally expressed proteins, but none of the known proteins contains more than 5 CUB domains. The extensive repetition of CUB domains provides another indication that this ~460 KDa is a multiligand receptor with multiple potential binding sites. Except for the leader peptide, no sequence compatible with a transmembrane domain or gchosylphosphatidylinositol (GPI) anchor-addition 17 signal could be identified in cubilin. This excludes the possibility that the protein is a transmembrane protein or GPI anchored protein that is synthesized with a cleavable hydrophobic C terminus. Early studies with guinea pig ileum homogenate without detergent treatment showed the receptor is soluble and maintained lF-Cbl binding activity (Cotter et al., 1976). Later study confirmed that association between the cubilin and membranes is weak since approximately 50% of cubilin could be released into the fluid phase during simple mechanical grinding without detergent or during incubation of membranes with heparin or EDTA (Moestrup et al., 1998). All these data suggested that cubilin is a peripheral membrane protein. As a peripheral membrane protein without internalization signals, how cubilin can facilitate the endocytosis of lF-Cbl complex is an open question. Co- internalization of a receptor that lacks internalization signal(s) by means of another receptor has been shown previously. Thus, the gchosylphosphatidylinositol-anchored urokinase receptor is endocytosed by coupling of urokinase receptor-bound urokinase-inhibitor complex to low density Iipoprotein receptor-related protein (LRP) (Nykjaer et al., 1997, Herz et al., 1992). Megalin, which is a closely related to LRP (both are low-density Iipoprotein receptor family members), also binds the urokinase-inhibitor complex (Moestrup et al., 1993), and may perform a similar function in regard to both the urokinase receptor and cubilin-ligand complexes. In vitro study showed high affinity interaction (Kd~7nM) between cubilin and megalin which does not interfere with subsequent lF-Cbl binding to cubilin (Moestrup et al., 1998; Hammad et al., 2000). In vivo immunohistochemistry of kidney, intestine and yolk sac in rat showed colocalization of these two proteins in these tissues (Sahali et al., 1988, 1993, Elm et al., 1997, Moestrup et al., 1998). Megalin has a single transmembrane domain and cytoplasmic domain motifs that signal for coated pit- mediated internalization. These data suggest that megalin might mediate cubilin endocytosis and recycling to the plasma membrane. Since the in vitro cubilin- megalin complex is stable at pH 5, the two receptors might remain in complex during the entire recycling pathway (Fig 1.3). Megalin IF-Cbl IFR iiiil till Plasma membrane PXY Figure 1.3 Schematic representation of the interaction between peripheral membrane protein cubilin and transmembrane protein megalin. Functional domains of cubilin To better understand the relationship between cubilin function and its structure, a mammalian expression approach has been used to identify the cubilin regions important for membrane association and ligand binding (Kristiansen et al., 1999). The results showed the N-terminal cubilin region is crucial for membrane association. This region has no classical transmembrane segment, but helical plotting demonstrated a conserved amphipathic helix motif that may be involved in the assembly of cubilin (rat amino acid 74-109, human amino acid 73-108). A recent electron microscopic study of purified bovine cubilin has shown the formation of trimers in solution without detergent or lipid (Lindblom et al., 1999). A region with four-heptad repeats, actually a part of the putative amphipathic helix structure, has been proposed to account for the assembly. The only cysteine located in the 110-amino acid N-terminal sequence, outside of the CUB domains and EGF repeats, might also account for the partial disulfide bond-dependent dimerization of a minor part of purified receptors. The binding site for lF-Cbl has been located in the region encompassing CUB domains 5-8 (Kristiansen et al., 1999). This establishes the CUB domain structure as a ligand-binding domain. The two mutations identified in the human cubilin gene both relate to the critical region now known to harbor the lF-Cbl binding region. The FM2 mutation, suggested to activate a cryptic intronic splice site leading to truncation of the receptor in CUB domain 6, may lead to an inactive or unstable translation product, whereas the P/L substitution (FM1) in CUB domain 8 might cause a structural change specifically impairing lF-Cbl binding. The CUB domain is a barrel-like structure containing two layers of five- stranded B-sheets with the B-turns in a surface-exposed position as in the antigen binding regions of immunoglobulins (Menendez et al., 1995; Dias et al., 1997). In view of the existence of 27 CUB domains in cubilin, a high number of 20 putative sites for various protein interactions may then be predicted. A site for binding of RAP was localized within CUB domains 13-14. The physiological importance of this interaction remains to be determined, but it is tempting to speculate that RAP may assist the processing/folding of cubilin in line with its suggested chaperone function which is important for normal processing of the LDL receptor-related protein and megalin (Willnow et al., 1995). RAP, a 40-kDa endoplasmic reticulum protein, serves as a novel kind of chaperone or escort protein and protects multiple ligand binding sites of processed low density lipoprotein receptor family proteins, in particular the two giant receptors megalin and LRP. The association with RAP indirectly indicates that cubilin is a multiple ligand binding protein. Regulation of cubilin expression in small intestine: Most gastrointestinal functions are not evenly distributed along the intestinal tract. The remarkable degree of coordination between the development of various aspects of gastrointestinal function suggests that the process may be triggered by a single or a few central mechanisms, such as weaning and (or) hormones (Hermiston and Gordon, 1995). Early studies showed the gradient of the enzymatic activity, or even the absorptive capacities for sugar and amino acids, correlated with the gradient of mucosa mass of the small intestine (Laganiere et al., 1984). However, later studies on the regulation of sucrase isomaltase (SI) and aminopeptidase expression along small intestine showed the translational or post-translational regulation of these enzymes, but no change of the mRNA level (Hoffman and Chang, 1991; Danielsen et al., 1986). Different 21 genes show different regional specificity and are under different regulatory mechanisms. During fetal development of the human intestine, cubilin activity can be detected along the entire length of intestine in early fetal development, but is located to the distal ileum by the end of fetal development (Schohn et al., 1992). Studies in rats also suggested that cubilin expression was developmentally regulated (Ramasamy et al., 1989; Ramanujam et al., 1993b). The functional expression of cubilin in distal small intestine was confirmed by oral administration of radio-labeled Cbl in dogs and humans (Baker et al., 1958; Fleming et al., 1962; Hagedorn and Alpers, 1977). The results of surgical transposition of ileal segments from distal to proximal part of the canine intestine further confirm the ileal location for lF-Cbl absorption in dogs (Drapanas et al., 1963). The distribution of cubilin along the gastrointestinal tract is against the gradient of the mucosa mass for the dog (Laganiére et al., 1984). The regional specific expression of cubilin along the small intestine must be regulated at some level. There is some in vivo evidence to suggest that the ileal capacity for Cbl absorption is regulated by modulation of cubilin expression during pregnancy (Brown et al., 1977; Robertson and Gallagher, 1983), but whether it is regulated by certain hormones needs to be investigated. From another point of view, the lining epithelium of the small intestinal mucosa is in a constant and rapid state of renewal (Weinstein WM, 1974; Leblond CP, 1980; Stappenbeck et al., 1998). Newly formed enterocytes arise in the crypt region and migrate up the villus to be shed into the intestinal lumen at 22 or near the villus tip. The overall process of cell renewal depends on the number of cells of the proliferative compartment; the rate of cell proliferation; and the migration time. During this migration, enterocytes mature and differentiate for a host of specific functions. Immunoelectrophoretic studies on human small- intestinal brush-border proteins showed the amounts of lactase, sucrase, maltase, microvillus aminopeptidase and dipeptidyl peptidase IV from jejunum biopsies had their maximum activities near the mid-region of the villi and their lowest activities at the base of the crypts (Skovbjerg H, 1981 ). How different gene expression along crypt to villus axis is regulated is still an unsolved question. One of the most well characterized gene regulatory mechanisms along the crypt-villus axis is the SI gene for which transcriptional regulation at the promoter depends on at least two groups of transcriptional proteins: hepatocyte nuclear factor 1(HNF-1) and caudal-related homeodomain proteins (Cdx)(Wu et al., 1994; Suh et al., 1996). An immunoelectron microscopic study (Levine et al., 1984) of canine cubilin distribution during enterocyte maturation from crypt to villus tip of ileal mucosa showed that synthesis of cubilin can be easily detected in the mid-villus cells, with the majority distributed intracellularly. Expression of cubilin dramatically increases in the villus tips, where also the majority of cubilin can be detected in the microvillus pits. The expression of cubilin is extremely low in the crypt cells. Later study (Kapadia and Essandoh, 1988) with enriched villus and crypt cell populations also showed IF-Cbl binding was about 30-fold greater in ileal villous cells than in crypt cells. From the above, it is evident that during cell 23 differentiation from crypt to villus, cubilin expression is up regulated during differenciation (maybe just slow). Although the particular signals affecting cubilin expression remain unknown, we can evaluate normal cubilin expression in different tissues, different regions of the same tissue, and the same region at different stages of cell differentiation to determine how cubilin expression is regulated. This will provide the basis for further studies of cubilin regulation and functions. Protein folding in the secretory pathway Being a membrane-associated protein, cubilin, like other secretory and membrane proteins, is likely carried to the plasma membrane via a specific membrane protein biosynthetic transport pathway. The first and most important compartment of this pathway is the endoplasmic reticulum (ER) which is responsible for protein synthesis, early posttranslational modifications, maturation and export of the fully folded protein to the destined location. Among all of these functions of ER, one of the most notable are mechanisms to monitor the fidelity of early biosynthetic events in the protein export pathway, also known as "ER quality control" (Kopito RR, 1997). This quality control is critical for normal cell function since it prevents premature export of incompletely or improperly folded proteins from ER. In recent years, numerous inborn errors of metabolism have been identified which affect secretory or plasma membrane proteins (Medeiros-Neto et al., 1996; Repaske et al., 1996; Kuivaniemi et al., 1991; Lehrman et al., 1987; 24 Yang et al., 1993). In many instances, mutations causing minor changes in proteins that result in a folding defect lead to abnormal protein trafficking, suggesting that proper conformation is critical for protein transport as well as biological activity. During protein biogenesis, a peptide chain can self-assemble into a stable, low free-energy conformation based on information contained within the primary structure. For many proteins, additional factors are required for proper folding. All these factors reside within the ER and are known as ER chaperones. According to their specific functional mechanism, some of them have been defined as primary controls and apply to all newly synthesized proteins. The others, defined as secondary controls, are specific for a particular protein or protein family (Ellgaard et al., 1999). The synthesis of primary molecular chaperones, including Bip, GRP94, and calnexin, etc., can be induced in response to the accumulation of unfolded secretory proteins (Chapman et al., 1998). This induction can be at the transcriptional level or posttranslational level. Because ER chaperone associations are based on recognition of features enriched in incompletely folded versions of exportable proteins, the Ca2+ and ATP dependent associations of ER chaperones are usually at their highest levels immediately upon nascent chain translocation into the ER, and terminated before export of the folded proteins from their compartment (Sambrook JF, 1990; Clairmont et al., 1991 ). The fact that molecular chaperones act on “substrate” proteins does not affect self- assembly. By interacting with nascent chains, chaperones prevent undesirable 25 protein-protein interactions and increase the chances that newly made proteins will have the opportunity to achieve their native structure. Primary control also includes disulfide isomerase and prolyl isomerase. They are the true foldases, in that they catalyze thioI-disulfide interchange with broad substrate specificity (Freedman, 1995). As the nascent chain enters the ER lumen, the rapid interaction within the hydrophobic domains is accompanied by the ordered formation of intramolecular disulfide bounds, which stabilize secondary and tertiary structures and can be critical for maintaining a biologically active conformation. Vlfithin the oxidizing ER environment, reactive thiols also can form mispaired disulfide bonds, and subsequent correction of aberrant disulfide bonds may represent one of the rate-limiting steps in protein folding. Specific cysteine residue mutations can also cause severe disulfide bond misformation which eventually blocks protein transportation (Reddy and Corley, 1998). Another important primary control mechanism involves oligosaccharide trimming (Parodi AJ, 1999). A 14-saccharide unit, which contains two N-acetyl glucosamine (GlcNac) residues, nine mannoses and three glucose resides, is initially added to asparagine-linked consensus acceptor sites (NXS/T) in glycoproteins. During ER enzyme-mediated trimming, the interaction between nascent proteins and ER chaperones (calnexin) are regulated by trimming enzymes, such as glucosyltransferase and glucosidase (Trombetta and Parodi, 1992; Sousa et al., 1992), which serve as folding sensors to direct the unfolded glycoprotein to ER chaperones. Thus, proper N-linked glycosylation and oligosaccharide trimming are also important for nascent protein folding. 26 Quite a number of protein specific factors that are responsible for effecting folding and assembly of nascent proteins have been identified in different species (Ellgaard et al., 1999). A well-studied factor, classified as an escort protein is receptor associate protein (RAP), the function of which was first recognized by its interaction with LRP molecules through the whole biosynthetic pathway, even remaining associated on the cell surface (Bu et al., 1995; Farquhar et al., 1995). Later study showed that RAP can also interact with other membrane receptors, like megalin (Moestrup et al., 1996) and cubilin (Moestrup et al., 1998). Absence of RAP can cause the defective transport of LRP through the ER (Willnow et al., 1995). With RAP association, both RAP and LRP can exit the ER, which suggests that the interaction maintains a favorable conformation and prevents premature association of LRP ligands in the ER, which could lead to receptor retention and [or degradation. RAP dissociation occurs when LRP reaches the cell surface and LRP ligands can displace the escort. Surface expression of RAP has been questioned in a study which demonstrated that RAP relocates to RAP binding site in vitro when tissues are snap frozen (Abbate et al., 1993). Cubilin is a large protein which contains 76 disulfide bonds and 47 potential N-glycosylation sites in human (Kozyraki et al., 1998). The assembly process within the ER must be time consuming and require a lot of ER chaperones. The in vitro interaction with RAP indicates that RAP or other proteins may act as chaperones to facilitate the intracellular maturation. It is not known whether cubilin-specific chaperones are involved in its processing. 27 Biochemical studies of affected dogs with selective intestinal Cbl malabsorption demonstrated the accumulation of cubilin in the ileal enterocytes, but rapid degradation in the kidney epithelial cells (Fyfe et al., 1991b, Fyfe, unpublished data). The different fates of the same protein in different tissues suggests that there is tissue-specific machinery for the protein assembly and export. The mechanism of defective expression of cubilin on the apical brush- border membrane of both ileal and kidney epithelial cells of affected dogs with selective intestinal Cbl malabsorption is the major focus of this project. The most common reason for an ER export disorder is mutation in the coding sequence of the exportable protein, eventhough defects of other accessory proteins may also be involved. The latter case has been demonstrated only in abetalipoproteinemia (Wetterau et al., 1992), a defect of the apo B-100 specific processing enzyme, microsomal triglyceride transfer protein. General outline for this thesis A dog family with selective intestinal cobalamin malabsorption, which closely resembles the human Imerslund-Grasbeck syndrome, provides a valuable genetic model to further investigate the mechanism of this inherited disease. The general goals of this thesis were to clone the candidate disease- causing gene, cubilin / intrinsic factor-cobalamin receptor (IFCR), and characterize its gene product. To achieve these goals, three closely related projects were conducted and are described in this thesis: (1) Cloning of the full- 28 length cDNA of canine cubilin, providing the molecular basis for further study of this candidate gene; (2) Studies on cubilin expression and post-translational modification in the canine gastrointestinal tract, which helped to better understand this newly cloned gene and the relationship between this gene and the disease in the dog family; and (3) Linkage analysis between the disease locus and candidate gene loci, which efficiently allowed determination of whether two candidate loci habored a disease-causing mutation. 29 References Abbate M, Bachinsky D, Zheng G, Stamenkovic I, McLaughlin M, Niles JL, McCluskey RT, and Brown D. (1993) Location of gp330/a2-m receptor- associated protein (az-MRAP) and its binding sites in kidney: distribution of endogenous az-MRAP is modified by tissue processing. EurJ Cell Biol 61: 139- 149. Abdelaal MA and Ahmed AF. (1991) Case report: Imerslund-Grasbeck syndrome in a Saudi family. Acta Paediatr Scand 80: 1109-1112. Allen RH, Seetharam B, Podell E, and Alpers DH. (1978) Effect of proteolytic enzymes on the binding of cobalamin to R protein and intrinsic factor: In vitro evidence that a failure to partially degrade R protein is responsible for cobalamin malabsorption in pancreatic insufficiency. J Clin Invest 61: 47-54. Altay C, Cetin M, Gtimrflk F, Irken G, Yetgin S. and Laleli Y. (1995) Familial selective vitamin B12 malabsorption (Imerslund-Grasbeck syndrome) in a pool of Turkish patients. Pediat Hematol Oncol 12: 1 9-28. Altay C. and Cetin M. (1999) Vitamin Biz absorption test and oral treatment in 14 children with selective vitamin B12 malabsorption. Pediatr. Hematol. Oncol. 16(2):159-163. Aminoff M, Tahvanainen E, Grasbeck R, Weissenbach J, Broch H, and de la Chapelle A. ( 1995) Selective intestinal malabsorption of vitamin B12 displays recessive mendelian inheritance: assignment of a locus to chromosome 10 by linkage. Am J Hum Genet 57: 824-31. Aminoff M, Carter JE, Chadwick RB, Johnson C, Grasbeck R, Abdelaal MA, Broch H, Jenner LB, Verroust PJ, Moestrup SK, Chapelle A, and Krahe R. (1999) Mutations in CUBN, encoding the intrinsic factor-vitamin B12 receptor, cubilin, cause hereditary megaloblastic anaemia 1. Nat Genet 21: 309-313. Baker SJ, Mackinnon NL, and Vasudevia P. (1958) The site of absorption of orally administered vitamin B12 in dogs. Ind J Med Res 46: 812-817. Batt RM, and Horadagoda NU. (1989) Gastric and pancreatic intrinsic factor- mediated absorption of cobalamin in the dog. Am J Physiol 257: G344-G349. Batuman V, Verroust PJ, Navar GL, Kaysen JH, Goda FO, Campbell WC, Simon E, Pontillon F, Lyles M, Bruno J, and Hammond TG. (1998) Myeloma light chains are ligands for cubilin (gp280) Am J Physiol 275(R44): F246-F254. 30 Ben-Bassat I, Feinstein A. and Ramot B. (1969) Selective vitamin B12 malabsorption with proteinuria in Israel-«Clinical and genetic aspects. Israel J Med Sci 5:62-68. Bim H, Verroust PJ, Nexo E, Hager H, Jacobsen C, Christensen El, and Moestrup S. (1997) Characterization of an epithelial ~460-kDa protein that facilitates endocytosis of intrinsic factor-vitamin B12 and binds receptor- associated protein. J Biol Chem 272(42): 26497-26504. Birn H, Fyfe JC, Jacobsen C, Mounier F, Veroust PJ, Orskov H, Willnow T, MoestrupSK, and Christensen El. (2000) Cubilin is an albumin binding protein important for renal albumin reabsorption. J Clin Invest (in press). Booth CC and Mollin DL. (1959) The site of absorption of vitamin B12 in man. Lancet 1: 18-21. Bork P and Beckmann G. (1993) The CUB domain. A widespread module in developmentally regulated proteins. J Mol Biol 231: 539-545. Bose S, Seetharam 8, Hammond TG, and Seetharam B. (1995a) Regulation of expression of transcobalamin Il-receptor in the rat. Biochem J 310: 923-929. Bose S, Seetharam S, and Seetharam B. (1995b) Membrane expression and interactions of human transcobalamin II receptor. J Biol Chem 270: 8152-8157. Broch H, Imerslund O, Monn E, Hovig T, and Seip M. (1984) Imerslund-Grasbeck anemia, A long-term follow-up study. Acta Paediatr Scand 73: 248-253. Brown J, Robertson J, and Gallagher N. (1977) Humoral regulation of vitamin B12 absorption by pregnant mouse small intestine. Gastroentenology 72: 881-885. Bu G, Geuze HJ, Strous GJ, and Schwartz AL.(1995) 39 kDa receptor- associated protein is an ER resident protein and molecular chaperone for LDL receptor-related protein. EMBO J 14: 2269-2280. Burman JF, Jenkins WJ, Walker-Smith JA, Phillips AD, Sourial NA, Williams CB, and MollinDL. (1985) Absent ileal uptake of IF-bound vitamin B12 in vivo in the Imerslund-Grasbeck syndrome (familial vitamin B12 malabsorption with protienuria). Gut 26: 311-314. Carmel R, Abramson SB, and Renner IG. (1983) Characterization of pure human pancreatic juice: cobalamin content, cobalamin-binding proteins and activity against human R binders of various secretions. Clin Sci 64: 193-205. 31 Celep F, Karagfizel A, Aynaci FM. and Erduran E. (1996) A case report of 46, XX, del (21) (q22) de novo deletion associated with Imerslund-Grasbeck syndrome. Clin Genet. 50:248. Chapman R, Sidrauski C, and Walter P. (1998) Intracellular signaling from the endoplasmic reticulum to the nucleus. Annu Rev Cell Dev Biol 14: 459-485. Christensen JM, Hippe E, Olesen H, Rye M, Haber E, Lee L, and Thomsen J (1973) Purification of human intrinsic factor by affinity chromatography. Biochim Biophys Acta 303, 319-332. Clairmont CA, De Maio A, and Hirschberg CB. (1992) Translocation of ATP into the lumen of rough endoplasmic reticulum-derived vesicles and its binding to luminal proteins including BiP (GRP 78) and GRP 94. J Biol Chem 267: 3983- 3990. Colle E, Greenberg L, and Krivit W. (1961) Studies of a patient with selective deficiency in absorption of vitamin B12. Blood 18: 48. Cooper BA and Castle WB. (1960) Sequential mechanisms in the enhanced absorption of vitamin 812 by intrinsic factor in the rat. J Clin Invest 39: 199-214. Cotter R and Rothenberg SP. (1976) Solubilization, partial purification and radioassay for the intrinsic factor receptor from the ileal mucosa. Br J Haematol 34: 477-487. Dan N and Cutler DF. (1994) Transcytosis and processing of intrinsic factor- cobalamin in Caco-2 cells. J Biol Chem 269(29): 18849-18855. Dias JM, Carvalho AL, Kolln l, Calvete JJ, Topfer-Petersen E, Varela PF, Romero A, Urbanke C, and Romao MJ. (1997) Crystallization and preliminary X- ray diffraction studies of aSF P, a bovine seminal plasma protein with a single CUB domain architecture. Protein Sci 6: 725-727. Dieckgraete BK, Seetharam B, and Alpers DH. (1988) Developmental regulation of rat intrinsic factor mRNA. Am J Physiol 254: G913-919. Donaldson RM, Mackenzie l, and Trier JS. (1967) Intrinsic factor-mediated attachment of vitamin B12 to brush borers and microvillous membranes of hamster intestine. J Clin Invest 46: 1215-1228. Doscherholmem A, Hagen PS, and Olin L. (1959) Delay of absorption of radiolabeled cyanocobalamin in the intestinal wall in the presence of intrinsic factor. J Lab & Clin Med 54: 434-439. 32 Drapanas T, Williams JS, McDonald JC, Heyden W, Bow T, and Spencer RP. (1963) Role of the ileum in the absorption of vitamin B12 and intrinsic factor (NF). JAMA 184: 337-341. Dugue B, Aminoff M, Aminoe—Gastin I, Leppanen E, Grasbeck R, and Gueant JL. (1998) A urinary radioisotope-binding assay to diagnose Grasbeck-Imerslund disease. J Pediatr Gastroenterol Nutr 26(1): 21 -25. El Kholty S, Guéant JL, Bressler L, Djalali M, Boissel P, Gerard P, Nicolas JP. (1991) Portal and biliary phases of entrohepatic circulation of corrinioids in humans. Gastroenterology 101: 1399-1408. Ellgaard L, Molinari M, and Helenius A. (1999) Setting the standards: Quality control in the secretory pathway. Science 286: 1882-1888. Farquhar MG, Saito A, Kerjaschki D, and Orlando RA. (1995) The Heymann nephritis antigenic complex: megalin (gp330) and RAP. J Am Soc Nephrol 6: 35- 47. Fenton WA and Rosenberg LE. (1989) Inherited disorders of cobalamin transport and metabolism. In Scriver CR, Beaudet AL, Sly WS, Valle D. (eds) The Metabolic Basis of Inherited Disease McGraw-Hill, New York. Festen HP. (1991) Intrinsic factor secretion and cobalamin absorption. Physiology and pathophysiology in the gastrointestinal tract. Scand J Gastroenterol 1 88 (supplement): 1-7. Fleming WH, King R, Galloway RA, and Roche JJ. (1962) The site of absorption of orally administered Com-labeled vitamin B12 in dogs: the effect of dose. Gastroenterology 42: 164-168. Freedman RB (1995) The formation of protein disulfide bonds. Curr Opin Struct Biol 5: 85-91. Fyfe JC, Jezyk PF, Giger U, and Patterson DF. (1989) Inherited selective malabsorption of vitamin B12 in giant schnauzers J Am Anim Hosp Assoc25: 533-539. Fyfe JC, Giger U, Hall CA, Jezyk PF, Klumpp SA, Levine JS, and Patterson DF. (1991a) Inherited selective intestinal cobalamin malabsorption and cobalamin deficiency in dogs. Pediatr Res 29: 24-31. Fyfe JC, Ramanujam KS, Ramaswamy K, Patterson DF, and Seetharam B. (1991b) Defective brush-border expression of intrinsic factor-cobalamin receptor in canine inherited intestinal cobalamin malabsorption. J Biol Chem 266: 4489- 4494. 33 Fyfe JC. (1993) Feline intrinsic factor (IF) is pancreatic in origin and mediates ileal cobalamin (cbl) absorption. J Vet Intern Med 7: 133 (abstract). Gordon Jl. (1989) Intestinal epithelial differentiation: new insights from chimeric and transgenic mice. J cell Biol 108: 1187-1 191. Grasbeck R, Gordin R, Kantero I, And Kuhlback B. (1960) Selective vitamin B12 malabsorption and proteinuria in young people. Acta Med Scand 167:289-296. Guéant JL, Masson C, Schohn H, Girr M, Saunier M, and Nicolas JP. (1992) Receptor-mediated endocytosis of the intrinsic factor-cobalamin complex in HT 29, a human colon carcinoma cell line. FEBS 297(3): 229-232. Gullberg R. (1972) Vitamin B 12 -binding proteins in normal human blood plasma and serum. Scand J Haematol 9(6):639-647. Hagedorn CH and Alpers DH. (1977) Distribution of intrinsic factor-vitamin B12 receptors in human intestine. Gastroenterology 73: 1019-1022. Hall CA. (1975) Transcobalamin I and II as natural transport proteins of vitamin B12. J Clin Invest 56: 1125-1131. Hall CA, Green-Colligan PD, and Begley JA. (1985) Synthesis of transcobalamin II by cultured human hepatocytes. Biochim Biophys Acta 838(3): 387-389. Hakami N, Neiman PE, Canellos GP, and Lazerson J. (1971) Neonatal megaloblastic anemia due to inherited transcobalamin || deficiency in two siblings. N Engl J Med 285: 1163-1170. Hammad SM, Stefansson S, Twal WO, Drake CJ, Fleming P, Remaley A, Brewer HB Jr, and Argraves WS. (1999) Cubilin, the endocytic receptor for intrinsic factor-vitamin B(12) complex, mediates high-density Iipoprotein holoparticle endocytosis. Proc Natl Acad Sci U S A 96(18): 10158-63 . Hammad SM, Barth JL, Knaak C, and Argraves WS. (2000) Megalin Acts in Concert with Cubilin to Mediate Endocytosis of High Density Lipoproteins. J Biol Chem 275: 12003-12008. Herbert V and Castle GBJ. (1961) Divalent cation and pH dependence of rat intrinsic factor action in everted sacs and mucosal homogenates of rat small intestine. J Clin Invest 40, 1978-1983. Herz J, Clouthier DE, and Hammer RE. (1992) LDL receptor-related protein internalizes and degrades uPA-PAI-1 complexes and is essential for embryo implantation. Cell 71: 41 1-421. 34 Hoffman LR and Chang EB. (1991) Determinants of regional sucrase-isomaltase expression in adult rat small intestine. J Biol Chem 266: 21815-20 Hooper DC, Alpers DH, Burger RL, Mehlman CS, and Allen RH. (1973) Characterization of ileal vitamin B12 binding using homogeneous human and hog intrinsic factors. J Clin Invest 52: 3074-3083. ldriss JM and Jonas AJ. (1991) Vitamin B12 transport by rat liver lysosomal membrane vesicles. J Biol Chem 266: 9438-9441. Imerslund O. (1960) Idiopathic chronic megaloblastic anemia in children. Acta Paediat Scand Suppl 49: 1-115. Imerslund O and Bjornstad P. (1963) Familial vitamin B12 malabsorption. Acta Haematol 30: 1-7. Kapadia CR, Serfilippi D, Voloshin K, and Donaldson RM. (1983) Intrinsic factor- mediated absorption of cobalamin by guinea pig ileal cells. J Clin Invest 71 : 440- 448. Kapadia CR and Essandoh LK. (1988) Active absorption of vitamin B12 and conjugated bile salts by guinea pig ileum occurs in villous and not crypt cells. Digest Dis Sci 33: 1377-1382. Katz M and Cooper BA. (1974a) Solubilized receptor for intrinsic factor-vitamin B12 complex from guinea pig intestinal mucosa. J Clin Invest 54: 733-739. Katz M, Mehlman CS, and Allen RH. (1974b) Isolation and characterization of an abnormal human intrinsic factor. J Clin Invest 53: 1274-1283. Kojima T, Miyaishi O, Saga S, lshiguro N, Tsutsui Y, and Iwat. (1998) The retention of abnormal type | procollagen and correlated expression of HSP 47 in fibroblasts from a patient with lethal osteogenesis imperfecta. J Pathol 184(2): 212-8. Kolhouse JF and Allen RH. (1977) Recognition of two intracellular cobalamin binding proteins and their identification as methyl-malonyl-CoA mutase and methionine synthase. Proc Natl Acad Sci USA 74:921-925. Kopito RR. (1997) ER quality control: the cytoplasmic connection. Cell 88(4): 427-30. Kozyraki R, Kristiansen M, Silahtaroglu A, Hansen C, Jacobsen C, Tommerup N, Verroust PJ, and Moestrup SK. (1998) The human intrinsic factor-vitamin B12 receptor, cubilin: molecular characterization and chromosomal mapping of the 35 gene to 10p within the autosomal recessive megaloblastic anemia (MGA 1) region. Blood 91(10): 3593-3600. Kozyraki R, Fyfe J, Kristiansen M, Gerdes C, Jacobsen C, Cui S, Christensen El, Aminoff M, Chapelle A, Krahe R, Verroust PJ, and Moestrup S. (1999) The intrinsic factor-vitamin B12 receptor, cubilin, is a high-affinity apolipoprotein A-I receptor facilitating endocytosis of high-density lipoprotein. Nat Med 5: 656-661. Kristiansen M, Kozyraki R, Jacobsen C, Nexe E, Verroust PJ, and Moestrup SK. (1999) Molecular dissection of the intrinsic factor-vitamin B12 receptor, cubilin, discloses regions important for membrane association and ligand binding. J Biol Chem 274(29): 20540-20544. Kuivaniemi H, Tromp G, and Prockop DJ. (1991) Mutations in collagen genes: causes of rare and some common diseases in humans. FASEB J 5: 2052-2060. Kuovonen I. (1980) Solubilization of the pig ileal intrinsic factor receptor with papain treatment and studies on the solubilized receptor. Biochim Biophys Acta 626: 244-53. Kuovonen l and Grasbeck R. (1980) A simplified technique to isolate the porcine and human ileal intrinsic factor receptors and studies on their subunit structure. Biochim Biophy Res Comm 86: 358-364. LaFramboise R, Cooper BA, and Rosenblatt DS. (1992) Malabsorption of vitamin 812 from the intestine in a child with cbIF disease: evidence for lysosomal- mediated absorption. Blood 80: 291 -292. Laganiere S, Berteloot A, and Maestracci D. (1984) Digestive and absorptive functions along dog small intestine: comparative distributions in relation to biochemical and morphological parameters. Comp Biochem Physiol 79: 463- 472. Leblond CP. (1980) The life history of cells in renewing systems. Am J Anat 160: 1 14-158. Lehrman MA, Schneider WJ, Brown MS, Davis CG, Elhammer A, Russell DW, and Goldstein JL. (1987) The Lebanese allele at the low density lipoprotein receptor locus. Nonsense mutation produces truncated receptor that is retained in endoplasmic reticulum. J Biol Chem 262: 401-410. Levine JS, Nakane PK, and Allen RH. (1980) lmmunocytochemical localization of human intrinsic factor: the nonstimulated stomach. Gastroenterology 79: 493- 502. 36 Levine JS, Nakane PK, and Allen RH. (1982) lmmunocytochemical localization of intrinsic factor-cobalamin bound to the guinea pig ileum in vivo. Gastroenterology 82: 284-290. Levine JS, Allen RH, Alpers DH, and Seetharam B. (1984) lmmunocytochemical localization of the intrinsic factor-cobalamn receptor in dog-ileum: distribution of intracellular receptor during cell maturation. J Cell Biol 98: 11 11-1118. Le Panse S, Ayani E, Mulliez N, Chatelet F, Cywiner-Golenzer C, Galceran M, Citadelle D, Roux C, Ronco P, and Verroust P. (1994) Antibodies to the 280-kd coated pit protein, target of teratogenic antibodies, produce alterations in the traffic of internalized proteins. Am J Pathol 145: 1526-1536 . Le Panse S, Galceran M, Pontillon F, Lelongt B, van de Putte M, Ronco PM, and Verroust PJ. (1995) Immunofunctional properties of a yolk sac epithelial cell line expressing two proteins gp280 and gp330 of the intermicrovillar area of proximal tubule cells: inhibition of endocytosis by the specific antibodies. EurJ Cell Biol 67: 120-129 . Le Panse S, Ayani E, Nielsen S, Ronco P, Verroust P, and Christensen El. (1997) Internalization and recycling of glycoprotein 280 in epithelial cells of yolk sac. EurJ Cell Biol 72: 257-67. Li N, Seetharam S, Rosenblatt DS, and Seetharam B. (1994) Expression of transcobalamin ll mRNA in human tissues and cultured fibroblasts from normal and TC ll deficient patients. Biochem J 301:585-590. Lindblom A, Quadt N, Marsh T, Aeschlimann D, Morgelin M, Mann K, Maurer P, and Paulsson M. (1999) The intrinsic factor-vitamin B12 receptor, cubilin, is assembled into trimers via a coiled-coil alpha-helix. J Biol Chem 274: 6374-6380. Mackenzie IL and Donaldson RM. (1972) Effect of divalent cations and pH on intrinsic factor-mediated attachment of vitamin B12 to intestinal microvillous membranes. J Clin Invest 51: 2465-2471. Mackenzie IL, Donaldson RM, Trier JS, and Mathan VI. (1972) Ileal mucosa in familial vitamin B12 malabsorption. New Engl J Med 286: 1021-1025. Marcoullis G, Rothenberg SP, and Labombardi VJ. (1980) Preparation and characterization of proteins in the alimentary tract of the dog which bind cobalamin and intrinsic factor. J Biol Chem 255: 1824-1829. Marsden KA, Newman NM, and Marsden DE. (1979) Imerslund’s syndrome --- A case from Australia and review of the literature. Aust Paediatr J 15:49-52. 37 Medeiros-Neto G, Kim PS, Yoo SE, Vono J, Targovnik HM, Camargo R, Hossain SA, and Arvan P. (1996) Congenital hypothyroid goiter with deficient thyroglobulin. Identification of an endoplasmic reticulum storage disease with induction of molecular chaperones. J Clin Invest 98: 2838-2844. Menendez M, Gasset M, Laynez J, Lopez-Zumel C, Usobiaga P, Topfer- Petersen E, and Calvete JJ. (1995) Analysis of the structural organization and thermal stability of two spermadhesins. Calorimetric, circular dichroic and Fourier-transfomi infrared spectroscopic studies. Eur J Biochem 234: 887-896. Mellman IS, Youngdah-Turner P, \Mllard HF, and Rosenberg LE. (1977) Intracellular binding of radioactive hydroxocobalamin to cobalamin-dependent apoenzymes in rat liver. Proc Natl Acad Sci USA 74:916-920. Moestrup SK, Nielsen S, Andreasen P, Jorgensen KE, Nykjaer A, Roigaard H, Gliemann J, and Christensen El. (1993) Epithelial glycoprotein-330 mediates endocytosis of plasminogen activator-plasminogen activator inhibitor type-1 complexes. J Biol Chem 268: 16564-16570 Moestrup SK, Birn H, Fischer PB, Peterson CM, Verroust PJ, Sim RB, Christensen El, and Nexo E. (1996) Megalin-mediated endocytosis of transcobalamin-vitamin B12 complexes suggests a role of the receptor in vitamin B12 homeostasis. Proc Natl Acad Sci USA 93: 8612-8617. Moestrup SK, Kozyraki R, Kristiansen M, Kaysen JH, Rasmussen HH, Brault D, Pontillon F, Goda F, Christensen El, Hammond TG, and Verroust PJ. (1998) The intrinsic factor-vitamin B12 receptor and target of teratogenic antibodies is a megalin-binding peripheral membrane protein with homology to developmental proteins. J Biol Chem 273(9): 5235-5242. Nicolas JP and Guéant JL. (1995) Gastric intrinsic factor and its receptor. BaiII Clin Haematol 8(3): 515-531. Nykjaer A, Conese M, Christensen El, Olson D, Cremona O, Gliemann J, and Blasi F. (1997) Recycling of the urokinase receptor upon internalization of the uPAzserpin complexe. EMBO J 16: 2610-2620. Okuda K and Fujii T. (1977) Solubilization of the ileal receptor intrinsic factor- vitamin B12 comples in the rat. J Lab Clin Med 89: 172-180. Parodi AJ. (1999) Reglucosylation of glycoproteins and quality control of glyc0protein folding in the endoplasmic reticulum of yeast cells. Biochim Biophys Acta 1426(2): 287-95. 38 Quadros EV, Regec AL, Khan KM, Quadros E, and Rothenberg SP. (1999) Transcobalamin lI synthesized in the intestinal villi facilitates transfer of cobalamin to the portal blood. Am J Physiol 277(1 Pt 1): 6161-166. Ramanujam KS, Seetharam S, Ramasamy M, and Seetharam B. (1991a) Expression of cobalamin transport proteins and cobalamin transcytosis by colon adenocarcinoma cells. Am J Physiol 260(23): G416-G422. Ramanujam KS, Seetharam S, Dahms NM, and Seetharam B. (1991b) Functional expression of intrinsic factor-cobalamin receptor by renal proximal tubular epithelial cells. J Biol Chem 266: 13135-13140. Ramanujam KS, Seetharam S, and Seetharam B. (1992) Leupeptin and ammonium chloride inhibit intrinsic factor mediated transcytosis of [57Co]cobalamin across polarized renal epithelial cells. Biochem Biophys Res Comm 182: 439-446. Ramanujam KS, Seetharam S, and Seetharam B (1993a) Intrinsic factor- cobalamin receptor activity in a marsupial, the American opossum (Didelphis virginiana). Comp Biochem Physiol Comp Physiol 104: 771-775. Ramanujam KS, Seetharam S, and Seetharam B. (1993b) Regulated expression of intrinsic factor-cobalamin receptor by rat visceral yolk sac and placental membranes. Biochimi Biophysi Acta 1146: 243-246. Ramaswamy M, Alpers DH, Triuppathi C, and Seetharam B. (1989) Cobalamin release from intrinsic factor and transfer to transcobalamin ll within the rat enterocyte Am J Physiol 257: 6791-797. Rappazzo ME and Hall CA. (1972) Cyanocobalamin transport proteins in canine plasma. Am J Physiol 222(1): 202-206. Reddy PS and Corley RB. (1998) Assembly, sorting, and exit of oligomeric proteins from the endoplasmic reticulum. Bioessays 20: 546-554. Repaske DR, Summar ML, Krishnamani MR, Gultekin EK, Arriazu MC, Roubicek ME, Blanco M, Isaac GB, and Phillips JA 3rd (1996) Recurrent mutations in the vasopressin-neurophysin II gene cause autosomal dominant neurohypophyseal diabetes insipidus. J Clin Endocrinol Metab 81: 2328-2334. Retief FP, Gottlieb CW, and Herbert V. (1966) Mechanism of vitamin B12 uptake by erythocytes. J Clin Invest 45: 1 907-1 91 5. Robertson JA, and Gallagher ND. (1983) Increased intestinal uptake of cobalamin in pregnancy does not require synthesis of new receptors. Biochimi Biophysi Acta 757: 145-150. 39 Robertson JA and Gallagher ND. (1985a) In vivo evidence that cobalamin is absorbed by receptor-mediated endocytosis in the mouse. Gastroenterology 88(4): 908-12. Robertson JA and Gallagher ND. (1985b) Intrinsic facotr-cobalamin accumulates in the ileum of mice treated with chloroquine. Gastroenterology 89: 1353-1359. Rubin HM, Giorgio AJ, Macdonald RR, and Linarelli LG. (1974) Selective malabsorption of vitamin B12: Report of a case with metabolic studies. Am J Dis Child 127: 713-717. Sahali D, Mulliez N, Chatelet F, Laurent-Nnter C, Citadelle D, Sabourin JC, Roux C, Ronco P, and Verroust P. (1993) Comparative immunochemistry and ontogeny of two closely related coated pit proteins. The 280-kd target of teratogenic antibodies and the 330-kd target of nephritogenic antibodies. Am J Pathol142: 1654-1667. Sahali D, Mulliez N, Chatelet F, Dupuis R, Ronco P, and Verroust P. (1988) Characterization of a 280-kD protein restricted to the coated pits of the renal brush border and the epithelial cells of the yolk sac. Teratogenic effect of the specific monoclonal antibodies. J Exp Med 167: 213-218. Sambrook JF. (1990) The involvement of calcium in transport of secretory proteins from the endoplasmic reticulum. Cell 61: 197-199. Schohn H, Gueant L, Leheup B, Saunier M, Grignin G, and Nicolas JP. (1992) Intrinsic factor receptor during fetal development of the human intestine. Biochem J 286: 153-156. Seetharm B and Alpers DH. (1981) Isolation and characterization of the ileal receptor for intrinsic factor-cobalamin. J Biol Chem 256(8): 3785-3790. Seetharam B, Bagur SS, and Alpers DH. (1981) Isolation and characterization of proteolytically derived ileal receptor for intrinsic factor-cobalamin. J Biol Chem 257(1): 183-189. Seetharm B, Levine JS, Ramasamy M, and Alpers DH. (1988) Purification, properties, and immunochemical localization of a receptor for intrinsic factor- cobalamin complex in the rat kidney. J Biol Chem 263(9): 4443-4449. Seetharam S, Ramanujam KS, and Seetharam B. (1992) Synthesis and brush border expression of intrinsic factor-cobalamin receptor from rat renal cortex. J Biol Chem 267: 7421-7427. 40 Seetharam B, Christensen El, Moestrup SK, Hammond TG, and Verroust PJ. (1997) Identification of rat yolk sac target protein of teratogenic antibodies, gp280, intrinsic factor-cobalamin receptor. J Clin Invest 99(10): 2317-2322 Segrest JP, De-Loof H, Dohlman JG, Brouillette CG, and Anantharamaiah GM. (1990) Amphipathic helix motif: classes and properties. Proteins 8: 103-107. Sievens CJ. (1964) Megaloblastic anemia in a 3 year old child. Blood. 23: 547. Siggaard C, Rittig S, Corydon TJ, Andreasen PH, Jensen TG, Andresen BS, Robertson GL, Gregersen N, Bolund L, and Pedersen EB (1999) Clinical and molecular evidence of abnormal processing and trafficking of the vasopressin preprohorrnone in a large kindred with familial neurohypophyseal diabetes insipidus due to a signal peptide mutation. J Clin Endocrinol Metab 84:2933-41. Simpson KW, Morton DB, and Batt RM. (1989) Effect of exocrine pancreatic insufficiency on cobalamin absorption in dogs. Am J Vet Res 50: 1233-1236. Simpson KW, Alpers DH, De Ville J, Swanson P, Farmer S, and Sherding RG. (1993) Cellular localization and hormonal regulation of pancreatic intrinsic factor secretion in dogs. Am J Physio! 265: 178-188. Skovbjerg H. (1981) lmmunoelectriophoretic studies on human small-intestinal brush-border proteins -- Relation between enzyme activity and immunoreactive enzyme along the villus-crypt axis. Biochem J 193: 887-890. Smith A (1962) Veganism: A clinical survey with observations of vitamin B12 metalbolism. Br Med J 1: 1655-1659. Sousa MC, Ferrero-Garcia MA, and Parodi AJ. (1992) Recognition of the oligosaccharide and protein moieties of glycoproteins by the UDP- Glc:glycoprotein glucosyltransferase. Biochemistry 31: 97-105. Spurling CL, Sacks MS, and Jiji RM. (1964) Juverile pernicious anemia. New Engl J Med. 271:995-1003. Stappenbeck TS, Wong MH, Saam JR, Mysorekar IU, and Gordon JI. (1998) Notes from some crypt watchers: regulation of renewal in the mouse intestinal epithelium. Curr Opin Cell Biol 10: 702-709. Stones DK and Ferreira M. (1999) Imerslund-Grasbeck syndrome in an African patient. J Trop Pediatr 45(2): 106-107. Suh E-R, Chen L, Taylor J, and Traber PG. (1994) A homeodomain protein related to caudal regulates intestine-specific gene transcription. Mol Cell Biol 14: 7340-7351. 41 Sullivan LW. (1970) Vitamin B12 metabolism and megaloblastic anemia. Seminars Hematol 7: 6-22. Trombetta SE and Parodi AJ. (1992) Purification to apparent homogeneity and partial characterization of rat liver UDP-glucosezglycoprotein glucosyltransferase. J Biol Chem 267: 9236-9240. Vaillant C, Horadagoda NU, and Batt RM. (1990) Cellular localization of intrinsic factor in pancreas and stomach of the dog. Cell Tissue Res 260: 117-122. Weinstein WM. (1974) Epithelial cell renewal of the small intestinal mucosa. Med Clin N Am 58: 1375-1386. Wetterau JR, Aggerbeck LP, Bouma ME, Eisenber C, Munck A, Herrnier M, Schmitz J, Gay G, Rader DJ, and Gregg RE. (1992) Absence of microsomal triglyceride transfer protein in individuals with abetalipoproteinemia. Science. 258 (6): 999-1001. VVillnow TE, Armstrong SA, Hammer RE, and Herz J. (1995) Functional expression of low density lipoprotein receptor-related protein is controlled by receptor-associated protein in vivo. Proc Natl Acad Sci USA 92: 4537-4541. Wu GD, Chen L, Forslund K, and Traber PG. (1994) Hepatocyte nuclear factor 1a (HNF-1a) and HNF-1B regulated transcription via two elements in an intestine-specific promoter. J Biol Chem 269: 17080-17085. Yang Y, Ducos R, Rosenberg AJ, Catrou PG, Levine JS, Podell ER, and Allen RH. (1985) Cobalamin malabsorption in three siblings due to an abnormal intrinsic factor that is markedly susceptible to acid and proteolysis. J Clin Invest 76: 2057-2065. Yang Y, Janich S, Cohn JA, and Wilson JM. (1993) The common variant of cystic fibrosis transmembrane conductance regulator is recognized by hsp70 and degraded in a pre-Golgi nonlysosomal compartment. Proc Natl Acad Sci USA 90: 9480-9484. Yetgin S, Ozsoylu S, and Zamani VP. (1983) Imerslund-Grasbeck syndrome and generalized malabsorption. Turk J Pedia 25:193-196. Youngdahl-Turner P, Rosenberg LE, and Allen RH. (1978) Binding and uptake of transcobalamin II by human fibroblasts. J Clin Invest 61 2133-141 42 CHAPTER 2 CLONING OF CANINE CUBILIN 43 Introduction Gastrointestinal absorption of vitamin 812 (also called cobalamin,CbI) is essential for animals, including humans, to maintain normal hematopoiesis and integrity of the central nervous system. Cbl absorption is a complex process and requires many Cbl binding proteins. A defect of any one of these proteins can cause Cbl deficiency. Cubilin, previously known as intrinsic factor-cobalamin receptor (IFCR), plays a critical role in endocytosis of the lF-Cbl complex in distal small intestine and reabsorption of various proteins in the kidney. A family of dogs with selective intestinal Cbl malabsorption (Fyfe et al., 1989) has been identified that has similar clinical and laboratory features as seen in humans with a disorder known as Imerslund-Grasbeck syndrome (l-GS) (Grasbeck et al., 1960; Imerslund 0., 1960). The affected puppies exhibited failure of gastrointestinal absorption of oral radiolabeled Cbl. Immunoelectron microscopy and cell fractionation studies demonstrated a defect in cubilin expression on apical brush border membranes of ileum and renal proximal tubules, whereas other brush-border proteins were expressed normally (Fyfe et al., 1991a, b). Purified normal and affected dog renal cubilin showed similar migration on SDS-PAGE, but affected dog cubilin had abnormal proteolytic peptide profiles and the asparagine-Iinked oligosaccharides were endoglycosidase H-sensitive. The cubilin of affected dogs accumulated in ileal enterocytes but was rapidly degraded in the renal epithelial cells. All these findings suggested that affected dog cubilin did not fold properly and did not reach the mid-Golgi compartment of the biosynthetic pathway, most likely being 44 retained in the endoplasmic reticulum (ER). Many such so-called ER storage diseases which selectively inhibit cell surface expression of a plasma membrane or secretory protein have been described (Moolenaar et al., 1997; Medeiros-Neto et al., 1996; Repaske et al., 1996; Kuivaniemi et al., 1991; Lehrman et al., 1987; Yang et al., 1993). With the exception of abetalipoproteinemia (Wetterau et al., 1992), these disorders have been attributed to mutations in the coding sequence of the exportable protein. Rat (Moestrup et al., 1998) and human (Kozyraki et al., 1998) cubilin have recently been cloned and showed a unique tandem arrangement of 8 EGF domains and 27 CUB domains. Two independent mutations have been identified in CUBN in 17 Finnish families with IG-S (Aminoff et al., 1999). To further characterize the canine model with selective intestinal Cbl malabsorption and to understand the multiple functions of the gene, we cloned the canine cubilin cDNA from dog kidney proximal tubule epithelial cells and compared the canine cubilin sequence to human and rat sequences. 45 Materials and Methods Peptide sequencing of purified canine renal cubilin Canine renal cubilin was purified by lF-Cbl affinity chromatography as previously described (Fyfe et al., 1991b) and transferred from SDS-PAGE gel to PVDF membrane. In situ endoLys-C or trypsin digestions, separation of peptides by microbore HPLC, and peptide sequencing were performed by the Protein Chemistry Facility of the Worcester Foundation for Experimental Biology (Shrewsbury, MA). Canine kidney cDNA library screening A canine CDNA library was constructed from RNA isolated from normal canine pooled kidney proximal tubular epithelial cells, using Trizol (Life Technologies, Bethesda, MD) according to the manufacturer’s instructions. Total RNA was sent to Stratagene (La Jolla, CA) for construction of the library in the bacteriophage lambda vector, Lambda Zap ll, using both random octamer and oligo dT priming of the cDNA synthesis reaction. This library was screened with a digoxigenin-Iabeled probe derived from the rat cubilin cDNA (a kind gift of Pierre Verroust, INSERM, Pairs, FR). Hybridizing plaques were purified, and the 1.6 kb cDNA insert of one (DX2) was sequenced, revealing 85% identity with the human cubilin sequence. DX2 was labeled with [32P1a-dATP to high specific activity using a random primer labeling kit (Boehringer Mannheim) and used as a probe to hybridize to 6 HybondTM-N nylon membrane (Amersham Pharmacia Biotech) lifts totaling 3X105 pfu from the canine kidney proximal tubule epithelial cDNA 46 library. Duplicate membrane sets were prehybridized in 6X SSC, 0.5% SDS, 5X Denhardts solution, 20mM NaH2P04, and 100 jig/ml sonicated sperm DNA for 4 hrs at 65°C. Hybridization was performed in the same solution after adding the denatured probe for 16 hours at 65°C. Membranes were washed twice with 6X SSC at room temperature and twice with 2XSSC and 0.5% SDS for 30 min at 68°C. Positively hybridizing plaques were picked and rescreened using the same probe for three more rounds. The true positive plaques were purified, and in vivo excision of the phagemid pBluescript (SK-) was performed according to Stratagene protocol. XL1-Blue bacteria were infected with the positive phagemids and grown on LB agar plates containing 50 pg/ml ampicillin. Plasmid DNA was purified from overnight MR 2000 cultures (MacConnell Research, San Diego CA) using QIAGEN’s plasmid purification kit (QIAGEN Inc. Santa Clarita CA). PCR amplification of cubilin cDNA probes for sequential screening: Sequence information obtained from the cDNA clones was used to design primers, F3922, 5’-CAGGCAACAACTGGCAACACGGT-B’, R4907, 5’- GCAGGGAACAATGGAGAGGATA-3’, for use in PCR reactions to amplify a cDNA probe for sequential rounds of screening. The fifty pl final reaction volume contained: 1X PCR buffer (Gibco BRL), 0.25 mM each dNTP, 2 mM MgClz, 0.25 uM each primer, 0.4 ng plasmid DNA template and 2.5 units Taq DNA polymerase (Gibco/BRL). After a hotstart denaturation at 95°C for 5 min, the reactions were carried out for 10 cycles of denaturation at 94°C for 1 min, 47 annealing at 62°C for 1 min, and extension at 72°C for 5 min. Then the annealing temperature was decreased to 58°C for 26 more cycles. The amplified fragments were size fractionated by agarose gel electrophoresis in 1X TAE and were gel purified to use as probes for the next round of screening. The screening was carried out using the same protocol and same amount of cDNA library as above. For a third round of screening, 5 micrograms of canine kidney cortex total RNA were reversed transcribed with oligo dT primer (Gibco) at 42°C to synthesize first strand cDNA. 2 pl of the RT product was used as template to amplify a cDNA probe using primers F5404, 5’-GGTCACTTGGTGGGGC- GATACT-3’ and R6066, 5'-GTCCAGGGAAAGGATGTTGAGTT-3’ designed from the 3’ end sequence of a newly cloned fragment. The PCR reactions were performed under the same conditions as above for 31 cycles except using 67°C annealing for 203 and 72°C extension for 2 min. The PCR product was gel purified and used as probe to screen the canine cDNA library again. In multiple rounds of screening with newly isolated insert cDNAs as probes in each round, 10 clones (DX2-DX11) containing overlapping inserts were selected and partially or fully sequenced on both stands. Most were sequenced by primer walking, but DX11, containing an ~5 kb insert was sequenced using an in vivo transposon-based kit for random insertions of primer sites (GPS-Genome Priming System, New England Biolabs, Beverly, MA). All sequences were generated by dideoxy termination cycle sequencing methods on an ABI 373A Automated Sequencer (Applied Biosystems, Inc, Foster, CA). cDNA sequences were assembled and analyzed using DNASTAR (DNASTAR, Madison, WI) or 48 SEQUENCHER software (Gene Codes Corp, Ann Arbor, MI) and have been submitted to GenBank (accession #AF137068). Position 1 of the canine cubilin cDNA refers to the first nucleotide of the full-length cloned cDNA. By this system, the A residue of the first ATG is at position 74. 49 Results Cloning of Canine Cubilin cDNA Using a single rat cubilin cDNA fragment to screen a kZap cDNA library derived from dog kidney proximal tubule epithelial cells, an initial 1.6 kb clone encoding a portion of canine cubilin was identified. Two independent clones which covered upstream and downstream sequences were identified using this 1.6 kb fragment as probe to screen the same canine cDNA library. A number of other clones were identified using polymerase chain reaction-generated probes for further screening. Fig. 2.1 schematizes five overlapping clones used to construct the final cDNA. Canine cubilin cDNA contains11,282 bp including 73 bp of 5’ and 349 bp of 3’ untranslated sequence. There was a poly A addition signal sequence 320 bp 3’ of the stop codon and 21 bp 5’ of the beginning of the poly A tail. The nucleotide sequence was 85 % identical to the human and 74 % identical to the rat cubilin sequences in GenBank. Primary structure of canine cubilin: The open reading frame (ORF) of 10,860 bp had a deduced amino acid sequence of 3,620 residues representing a 397,435 Da protein (Fig. 2.2). Alignment of the deduced amino acid sequence demonstrated overall amino acid identity of 83 % with human and 70 % with rat cubilin. The primary structure of canine cubilin, which contains a 110 amino acid N-terminal sequence; 8 EGF 50 .xcmmcow 5 30533.0. 5.53 am. 9: 9 .8352 ea: new :25: 9: 9 30:52 e\emm mm; 8538 258.03: of. .33 < boa 2: Co @5563 9: Co .m an em ucm :88 noun 9.: Go .m an own 8533 .956 .5525 < 23 m mm; 99:. .8533 8529.5 .m ho an mvm ucmb ho an MK mEmEoo 95.0 <200 of. .NmNF Tvvmm H3x0 cam ovmuoémmm H85 ”334.58 ”38.33 Uaxe Hmmomémxxwxnu 80:20 02» >9 omnooco 8:303 (200 an Nam. E. on... 6:33 >963 ocEno ho m:_:o_o 400 kDa (labeled IC forms). The band observed at high apparent Mr (labeled ?) was also observed in canine renal cortex (Fig 3.4, lanes 4 and 5). None of these 4 cubilin species was observed when the immunoprecipitation or immunodetection on western blots was performed with nonimmune serum (not shown). Thus, small intestinal expression of immunospecific cubilin corresponded to expression of cubilin ligand-binding activity. 78 Cubilin mRNA expression in multiple segments of gastrointestinal mucosa was examined in each dog by northern blot (Fig 3.20). As demonstrated in Fig 3.1, no hybridizing CUBN transcripts were observed in stomach (St) or colonic mucosa. Hybridization of the cubilin probe to mucosal RNA was observed only in samples isolated from the distal half of small intestine, and highest expression was found in mucosa at 85-95% of the distance from pyloroduodenal to ileocolic junctions. In all samples in which hybridization of the probe was observed, a strongly hybridizing transcript at 11-12 kb and a more faintly hybridizing transcript at 13-14 kb were seen, just as in the multiple tissue blot. In order to compare the small intestinal distributions of the cubilin IF-cbl binding function and mRNA in a quantitative way, both sets of data for each dog were expressed as fractions of the peak activity observed (Fig 3.3). In the proximal to distal direction, lF-cbl binding (upper panel) and cubilin mRNA (lower panel) in each dog were first detectable in mucosa at 45-50 % and both parameters were maximal at 85-90 % of the small intestinal length. Regression analysis of the combined data of all 5 dogs in the interval from first detectable to maximal indicated that both parameters increased linearly (slopes of 2.1 and 2.0, respectively) with comparable variance (R2 values of 0.82 and 0.78, respectively). Thus, in this group of dogs, cubilin function and mRNA expression in the small intestine were highly correlated, both in distribution of detectable expression and the position of maximal expression. 79 Fraction of Peak IF-cbl Binding Activity Fraction of Peak mRNA Level I 20 40 60 80 100 Figure 3.3: Codistribution of quantitated intestinal cubilin ligand-binding activity and mRNA. Top panel. The IF-cbl binding specific activities represented in Fig 3.2A, were recalculated as fractions of the peak value in the intestine of each dog. Linear regression of the data between the first nonzero values and the peak values of all 5 dogs had a correlation coefficient of 0.82. Bottom panel Relative cubilin mRNA hybridization in each mucosal sample was determined by exposure of northern blots to a storage phosphor screen. The data are plotted as fractions of the peak value found in each dog. As in the top panel, linear regression of the data between the first nonzero values and the peak values of all 5 dogs had a correlation coefficient of 0.78. 80 Ileal cubilin membrane association To determine whether endogenous cubilin in ileal mucosa behaves as a weakly bound peripheral membrane protein in vitro, as has been reported for renal cubilin (Moestrup et al., 1998), we examined the distribution of cubilin after homogenization and fractionation of ileal mucosa without detergent. Ileal mucosa was homogenized in neutral hypotonic buffer, and an aliquot was centrifuged at 40,000 x g separating soluble and total membrane fractions. Apical microvillus brush-border membranes were enriched from the remaining homogenate by CaCIz aggregation of membranes followed by differential centrifugation, with recovery of brush-border membranes in the 16,000 x g pellet (Kessler et al., 1978). In a typical fractionation of 1 g of ileal mucosa, lF-cbl binding activity was 3.1 and 3.9 pmol (0.10 and 0.18 pmol/mg protein), in the soluble and total membrane fractions, respectively, and 0.59 pmol (0.41 pmol/mg) in the brush- border enriched fraction, indicating 2.7 fold enrichment of cubilin ligand binding over the homogenate and recovery of 8.4 % in the brush-border fraction. In contrast, enrichment of alkaline phosphatase (ALP) specific activity, a tightly associated apical brush-border membrane marker, was ~ 8.5 fold and recovery was ~ 45 % in the brush-border fraction. This suggested that some portion of cubilin may have been redistributed from the membrane to soluble fractions. In order to examine this more closely, lF-cbl binding proteins in each of the 40,000 x g soluble, total membrane, and brush-border-enriched fractions were recovered by Triton-X-100 solubilization and affinity chromatography. They were detected on western blots of nonreducing SDS-PAGE gels with polyclonal 81 rabbit anti-renal cubilin serum (Fig 3.4). Cubilin from the 40,000 x g supernatant and total membrane fractions are shown in lanes 1 and 2, respectively, and cubilin recovered from the brush-border membrane enriched fraction is shown in lane 3. The IF-cbl binding proteins recovered in parallel from canine kidney cortex homogenate are shown on the same gel in lane 4. The same single major band was detected in lane 5 by western blot of canine kidney homogenate without prior fractionation or affinity chromatography, indicating that the anti- cubilin serum used as primary antibody was also immunospecific in whole tissue homogenates. Three major cubilin bands were found in each ileal fraction (Fig 3.4). Two bands migrated just above and below the position of kidney cubilin with size estimates of 550 and 450 kDa, respectively (labeled IC), and a diffuse band migrated at ~185 kDa (labeled BB). There were also 2 faint bands of very high apparent Mr in each fraction (labeled ?). In 2 independent experiments, the proportion of total cubilin in each fraction represented by each size cubilin form was determined by densitometry of exposed films. The diffuse BB band comprised 30 % of recovered cubilin proteins in the soluble fraction, 55 % in the total membrane fraction, and 60 % in the brush-border enriched fraction, indicating an enrichment in the brush-border fraction of 1.5-2 fold relative to the total homogenate. In addition, the smaller 10 form in the brush-border fraction (lane 3) was depleted about 4 fold relative to that in the soluble fraction (lane 1) and about 2 fold relative to the total membrane fraction (lane 2). The larger IC 82 kDa ff} 4 ~460 p M 4 200 I d 116 Figure 3.4: lmmunoreactive and lF-cbl binding cubilin species in fractionated canine ileal mucosa and renal cortex. Cubilin species were partially purified by IF-cbl affinity chromatography from various non-detergent fractions of ileal mucosa (lanes 1-3) or renal cortex homogenate (lane 4) and separated by non-reducing SDS-PAGE on 5 % gels. Ileal mucosa fractions were 40,000 x g supernatant of total homogenate (lane 1), membranes pelleted at 40,000 x g from total homogenate (lane 2), and brush-border membranes enriched from homogenates by CaCl2 precipitation (lane 3). For comparison, immunoreactive cubilin of renal cortex homogenate without lF-cbl affinity purification is also shown (lane 5). Cubilin species were detected on western blots with polyclonal anti-canine renal cubilin serum and an anti-rabbit IgG- alkaline phosphatase conjugate, either by chemiluminescent exposure of autoradiographic film (lanes 1-4) or by incubation in alkaline NBT/BCIP solution (lane 5). form represented about 20 % of cubilin in all 3 fractions. The 145 kDa band was judged to be nonspecific crossreactivity because it was always present in the chemiluminescent detection system as the only band detected and no decrease was observed in signal on films of control blots developed with nonimmune rabbit serum as the primary antibody, with the anti-rabbit IgG-ALP conjugate only, or with [12511-protein A. Detection with the latter 2 control reagents suggested that the 145 kDa band represented crossreacting, nonreduced canine lgG from lymphocytes in ileal mucosa. The seeming enrichment of the very high apparent MIr form (?) in lane 3 was not a consistent finding and often occurred in the other fractions. Inclusion of disulfide bond reducing agents in the gel loading buffer in preliminary experiments resulted in an increased apparent Mr, but 10-20 fold reduction in signal, of renal cubilin and complete failure to detect ileal cubilin bands (Fig 3.5). The same set of 3 cubilin bands was also recovered from the final supernatant after pelleting the brush-border fraction at 16,500 x g. In order to examine whether cubilin had redistributed to a truly soluble fraction, ileal mucosa was again homogenized without detergent, but in neutral isotonic buffer without CaClz, and centrifuged at 40,000 x g for 1 hr, and the resulting supernatant was centrifuged again at 150,000 x g for 2 hr. IF-cbl binding proteins were affinity purified from each fraction and western blotted as above. All 3 of the cubilin forms identified above were present in each of these fractions, but there was no enrichment of the BB form or diminution of the smaller IC form apparent in any fraction (Fig 3.6). lmmunoreactive cubilin was roughly equally distributed 84 ’1"? Mi e— 200 kDa Figure 3.5: Western blot of canine cubilin in reducing and non- reducing condition. Canine cubilin purified by affinity chromatography from normal dog kidney (lanes 1 and 2) and normal dog ileum (lanes 3 and 4) was loaded with 100 mM DTT (lanes 1 and 3) and without DTT (lanes 2 and 4) onto 5% polyacrylamide gel. The proteins were detected by sequential incubation in polyclonal anti-canine renal cubilin serum, anti-rabbit lgG-alkaline phosphatase conjugate, and alkaline NBT/BCIP solution. 85 between the 150,000 x g supernatant and pellet fractions. Thus, none of the identified ileal cubilin forms appeared to be tightly membrane bound. Oligosaccharide analysis To further characterize the apparent size and tissue-specific differences of cubilin species, proteins purified from the 40,000 x g soluble fraction of ileal mucosa and renal cortex homogenates by IF-cbl affinity chromatography were subjected to differential glycosidase digestion prior to nonreducing electrophoretic separation and western blotting (Fig 3.7). Renal (lanes 1-3) and ileal cubilin (lanes 4-9) were digested with endo H (lanes 2, 5, and 8) or PNGase F (lanes 3, 6, and 9). Lanes 1, 4, and 7 were mock digestions with no enzyme added. The renal and ileal proteins in lanes 1-6 were from normal dog tissues, and the ileal proteins in lanes 7-9 were from mucosa of a dog affected with inherited selective cobalamin malabsorption due to failure of brush-border cubilin expression (Fyfe et al., 1991). There was nonspecific protein degradation apparent in both mock and real digestions of renal cubilin that was not apparent in digestions of ileal cubilin. Endo H digestion of renal cubilin shifted its migration only slightly, whereas PNGase F digestion reduced the apparent size to about 400 kDa. Endo H digestion of ileal proteins reduced the apparent size of the ~450 kDa band to ~400 kDa but had no affect on gel migration of the other major ileal cubilin species. In contrast, PNGase F digestion of ileal cubilin reduced the apparent size of both the ~550 and ~450 bands to ~400 kDa and the ~185 BB form to 145 kDa. As previously described (Fyfe et al., 1991), the BB form was 86 IC form ( BB form-O :3 Figure 3.6: Distribution of immunoreactive cubilin forms in different centrifugation fractions of dog ileal homogenate. Ileal mucosa was homogenized without detergent or CaCI2 (lane H), and centrifuged at 40,000 x g for 1 hr. The resulting supernatant (lane 8,) was centrifuged again at 150,000 x g for 2 hr to produce a final supernatant (lane 82) and membrane pellet (lane p2) lF-cbl binding proteins were affinity purified from each fraction and western blotted as described for Fig 3.4. 87 IEndo H IPNGaseF kDa ~550 ~450 ~400 ..,'.V.V 200 P. 116 D Figure 3.7: Oligosaccharide analysis of affinity purified ileal and renal cubilin. Renal cubilin of a normal dog (lane 1-3), ileal cubilin of a normal dog (lanes 4-6), and ileal cubilin of a dog affected with an inherited defect blocking brush-border expression of cubilin (lanes 7-9) were concentrated on IF-cbl- agarose beads. Eluted proteins were dialyzed and mock digested (lanes 1, 4, and 7), digested with endo H (lanes 2, 5, and 8), or digested with PNGase F (lanes 3, 6, and 9) prior to separation by non-reducing SDS-PAGE on 5 % gels. Proteins were detected by chemiluminescent western blot as described for Fig 3.4. 88 missing from affected dog ileal cubilin species, but the remaining species exhibited similar shifts in size as normal dog ileal cubilin after both glycosidase digestions. Cubilin expression along crypt-villus axis in dog ileum Cell fractions isolated from ileum during sequential intervals of incubation in EDTA represent cell populations of various stages of differentiation along the crypt to villus axis. The cell populations in early fractions contain mainly apical villus cells at a late stage of differentiation. With longer incubation, the cell populations encompass the villus base and crypt cells in early stages of differentiation (Weiser et al., 1973). Northern blot analysis of these sequential cell fractions, from two dogs, showed that the cubilin transcripts could be only detected in the first 4-5 fractions with the highest level expression occurring in the first or second fractions. In these two fractions, the majority of the cells were assumed to be villus tip cells. The expression levels gradually decreased after the fourth fraction. No cubilin transcript could be detected in the last 4 fractions. GAPDH control mRNA could be detected in all cell fractions (Figure 3.8). To confirm the reliability of the method of cell fractionation, the cell fractions from the same two dogs were tested for villus tip and crypt specific markers. Alkaline phosphatase (ALP) which is only expressed in the mature villus cells was used as a tip specific marker (Traber et al., 1992), and the canine homolog of mouse cryptidin related protein-ductin (CRP-ductin), which is only 89 Fraction 1 2 3 4 . 5 6 7 “8 9 Dog 1 cubilin ' CRP-ductin GAPDH GAPDH Figure 3.8: Northern blot analysis of canine cubilin and CRP-ductin along the crypt-villus axis in dog ileum. Thirty pg of total RNA isolated from dog ileal epithelial cell fractions were used for the northern blots and hybridized sequentially to dog cubilin cDNA, dog CRP-ductin cDNA amplified by RT-PCR, and loading control probe GAPDH. 90 Figure 3.9: Alkaline phosphatase activity along the crypt-villus axis in dog ileum. Total homogenate of dog ileal epithelial cell fractions were used for alkaline phosphotase (ALP) activity analysis after protein quantitation. The horizontal axis represents the cell fractions from villus to crypt. The vertical axis represents the ALP activity in U/mg protein. The solid and dashed line represent the duplicate experiment for the same dog. 91 unltlmg protein Unltlrng protein ALP activity In Ileal epItheIaII celh of 6091 0.1 0.09 __-_ _ 0.01 ——- -— .-....";a; fraction 1 traction 2 fraction 3 traction 4 fraction 5 fraction 6 fraction 7 fraction 8 fraction 9 cell fraction from vlllua to crypts ALP actIvIty In Ileal epithelial cells of dog 2 O1 008, 0% 0.04 002 , Fraction 1 Fraction 2 Fraction 3 Fraction 4 Fraction 5 Fraction 6 Fraction 7 Fraction 8 Fraction 9 Fraction 10 Fractions of Iaolated cells from villus to crypts 92 expressed in the immature crypt cells, was used as a crypt specific marker (Cheng et al., 1996). For ALP activity in each dog (Figure 3.9), the peak ALP activity was detected in the first fraction, and the activity decreased gradually in the subsequent fractions. These results suggested that the cell populations in the first three fractions represented mainly the villus tip cells, that cells in the middle three fractions represented a mixture of villus and crypt cells, and that cells in the last three fractions were mainly crypt cells. However, the same membrane rehybridized with the crypt specific probe (CRP-ductin) showed that the canine CRP-ductin transcript could be detected in almost all the fractions rather than just the last 4-5 fractions. High level expression of CRP-ductin was detected in fractions 2-3 for one dog and in fractions 3-5 for the other dog (Figure 3.8). These data did not support our assumption that the fractions with cubilin expression contained mainly villus tip cells. 93 Discussion Functional expression of a plasma membrane receptor may be regulated at one or more of multiple biosynthetic steps. The magnitude of changes in intestinal cubilin mRNA expression from segment to segment demonstrated here accounted fully for the observed changes in cubilin ligand-binding activity and immunodetectable protein, indicating that the most important molecular mechanism regulating regional distribution of cubilin expression in the canine gastrointestinal tract is modulation of cubilin mRNA levels. The sharp decline of both cubilin mRNA expression and ligand-binding activity in the most distal segment of small intestine was most probably due to sampling from areas of modified epithelium overlying a high density of Peyer's patches. The codistribution of cubilin function, immunoreactive protein, and mRNA expression correlated well with previous in vivo localizations of lF-cbl binding and absorption in canine small intestine by oral radiolabeled cbl administration (Baker et al., 1958; Drapanas et al., 1963; Fleming et al., 1962; Marcoullis et al., 1981) and is similar to that demonstrated by mucosal ligand binding measurements in humans (Hagedorn and Alpers, 1977). In the present study, however, we measured the IF-cbl binding function of cubilin in homogenates of whole mucosa and did not differentiate the activity of cell-surface receptor from that of intracellular cubilin. We cannot comment, therefore, on whether endocytic functions mediated by cubilin on the enterocyte surface may be regulated also by receptor translocation to the plasma membrane, as has been described for glucose transporters (Kahn, 1992), or by other post-translational mechanisms. 94 In examining cubilin mRNA expression by northern blot with a canine renal cubilin cDNA probe, two principal hybridizing transcripts were detected in distal small intestinal mucosa and renal cortex. The 11-12 kb transcript corresponds in size to the length of cloned renal CUBN cDNA (Xu et al., 1999) and hybridized ~ 4 fold more strongly than did the larger transcript. Because the blotted samples included nuclear RNA, the multiple hybridizing transcripts could represent mature cubilin mRNA and incompletely processed forms of the primary CUBN transcript still retaining one or more introns. As previously reported, Southern blots of canine DNA digested with several restriction endonucleases indicated only a single hybridizing CUBN locus in the canine genome (Xu et al., 1999), thus eliminating the possibility that we observed hybridization to multiple closely related transcripts of different genes. An alternative explanation which has not been examined is that the CUBN transcript may be alternatively spliced. The polyclonal antibody used in this study was raised against purified renal cubilin of a single apparent Mr but specifically detected 3 major proteins on western blots of ileal mucosal proteins. This indicated that the multiple immunoreactive forms of cubilin observed in ileal mucosa were not simply members of a protein complex obtained by co-immunoprecipitation or IF-cbl binding of one member of the complex. In preliminary experiments, it was determined that some form of receptor concentration, either immunoprecipitation or affinity chromatography, was necessary for reliable immunodetection of ileal cubilin on western blots. This may explain why cubilin was not detected on western blots of ileal mucosa from a pair of Imerslund-Grasbeck patients 95 purported to overexpress cubilin ligand binding activity (Eaton et al., 1998). The very high apparent Mr bands observed on western blots of ileal and renal cortex (labeled ? in Fig 2B and 4) were most likely aggregates of cubilin that survived boiling in SDS. We have observed these also on silver stained gels of affinity purified ileal and renal cubilin so they are not a western blotting artifact. The aggregate forms in ileal mucosa were higher apparent Mr than in renal cortex (Fig 3.4), and they shifted migration on SDS-PAGE after glycosidase digestions (Fig 3.7). Both observations were consistent with the more extensive N-Iinked glycosylation on the 550 kDa form of ileal cubilin than that of renal cubilin evident from the relative shifts in gel migration after PNGase F digestion (Fig 3.7). Heavy glycosylation of ileal cubilin may be an adaptation conferring some protection from Iumenal proteases, and may explain the greater stability of ileal vs renal cubilin apparent during glycosidase digestions in vitro. Physiochemical evidence of cubilin aggregation under various in vitro conditions has been reported previously (Birn et al., 1997; Kristiansen et al., 1999; Lindblom et al., 1999; Seetharam et al., 1981a; Seetharam et al., 1982). In a recent report, trimers of bovine renal cubilin of ~1,500 kDa were resistant to disruption in 6 M guanidine but were separated into 440 kDa monomer after disulfide bond reduction (Lindblom et al., 1999). The N-terminal region conjectured to mediate trimer formation has also been identified as a region required for membrane association of cubilin fragments expressed in transfected cells (Kristiansen et al., 1999). In the latter study, aggregate forms were observed on nonreducing gels only in the constructs expressing the membrane 96 binding N-terminal sequence. This region of cubilin also has the only unpaired cysteine in the entire protein that would be available for dimer formation. The presence of aggregate forms is, however, an inconsistent observation, so it remains unclear whether cubilin multimer formation occurs in vivo or is merely an in vitro artifact. We concluded that the ~185 kDa band, labeled BB in Fig 32B and 3.4, is a brush-border form of cubilin based on 1) its absence from ileal mucosa of dogs exhibiting an inherited defect that blocks brush-border cubilin expression and 2) its enrichment in fractions enriched for brush-border membranes. It is likely that the ~185 kDa form arises by proteolytic cleavage of the ~550 kDa form (see below) when exposed to proteases in the intestinal lumen. This idea is consistent with the previous finding by gel exclusion chromatography that in vitro trypsin treatment converted high molecular weight canine ileal cubilin to lF-cbl binding forms of 150-180 kDa (Seetharam et al., 1982). Enrichment and recovery of cubilin ligand binding activity and immunoreactivity of the ~185 kDa form was considerably less than that of the tightly-associated brush-border membrane marker, ALP, but it was clear that a portion of brush-border cubilin redistributed to other fractions during homogenization. This study confirmed that endogenous cubilin is only loosely membrane associated in canine ileum, similar to what was previously noted with endogenous cubilin in rabbit kidney (Moestrup et al., 1998) and canine ileal cubilin reconstituted in artificial liposomes (Seetharam et al., 1981 b). These are in vitro behaviors consistent with the designation of cubilin as a peripheral membrane protein. 97 What we have designated IC forms in Fig 3.2B and 3.4 appear to be intracellular cubilin precursors at different stages of maturation. The N-linked oligosaccharides of the smaller IC form, estimated to be 450 kDa, were cleaved by endo H, indicating that they are high-mannose and derived from cubilin which has not been transported from the RER to the cis-Golgi. Furthermore, the 450 kDa band was found largely in the 40,000 x g soluble fraction after homogenization in TMB without detergents and was depleted from the brush- border enriched fraction, consistent with an association with RER derived microsomes. Oligosaccharides of the larger IC form, estimated to be 550 kDa, were endo H resistant but PNGase F susceptible, indicating they are complex or hybrid types that had been exposed to Golgi processing enzymes. The same was true of oligosaccharides on the 185 kDa form. We have not determined which of these forms is directly responsible for lF-cbl endocytosis in vivo. The 185 kDa and 550 kDa forms may coexist for some time on the brush-border, and it is possible that the endocytic function of cubilin is inactivated by a proteolytic cleavage to 185 kDa. Detection of the ~450 kDa form was evidence that ileal cubilin gained IF- cbl binding activity and immunoreactivity at a step in biosynthesis prior to modification of high mannose oligosaccharides to endo H resistant forms and prior to expression in a position on the brush-border appropriate to its endocytic function. Previous data indicated that the same was true of renal cubilin (Fyfe et al., 1991). At the steady states of tissues examined in this report, renal cubilin was mostly endo H resistant, but a large proportion of ileal cubilin was endo H 98 sensitive. However, this does not reflect a true tissue-specific difference of cubilin partitioning between the RER and post-RER compartments because renal cells that express cubilin are largely a population of fully differentiated cells whereas ileal enterocytes that express cubilin in a homogenate of mucosa are a mixture of proliferating and immature crypt cells, partially differentiated mid-villus cells, and fully differentiated villus tip cells. Thus, in contrast to kidney, homogenates of ileal mucosa are relatively enriched in cubilin forms produced early in the biosynthetic pathway and the mature forms are underrepresented. Removal of N-linked oligosaccharides from both IC forms of ileal cubilin resulted in migration at ~400 kDa, consistent with the polypeptide molecular weight deduced from the canine renal cubilin cDNA (Xu et al., 1999). Similar to most glycoproteins destined for the plasma membrane, it appears that canine ileal cubilin is translated as a 400 kDa polypeptide, that it is N-Iinked glycosylated in the RER to ~450 kDa, and thereafter, that oligosaccharide modifications produce a mature ~550 kDa glycoprotein. However, as shown in Fig 3.7, dogs exhibiting an inherited defect blocking brush-border expression of cubilin still produce both IC forms of cubilin in the intestine, including the endo H resistant, ~550 kDa form. Genetic linkage evidence indicates that the disease locus in these dogs is not the cubilin locus (Xu et al., 1999), and therefore, some type of cubilin-specific transport to the Iumenal brush-border surface must exist. The present data suggest that it operates after cubilin matures to the endo H resistant, ~550 kDa form in ileal mucosa, but a study in rat yolk sac cells suggests that newly synthesized cubilin arrives on the brush-border with endo H 99 sensitive oligosaccharides (Baricault et al., 1995). It is possible that these phenomena are tissue or species specific. Tissue-specific differences of N-linked oligosaccharides may also have contributed to confusion in the literature about ileal cubilin protein size and lingering thought that intestinal and renal cubilin ligand-binding activity might be due to expression of different but closely related genes (Guéant et al., 1999). The present data indicated that ileal and renal cubilin in dogs have the same protein backbone and that differences of apparent Mr on SDS-PAGE are due to differing N-linked glycosylation. Moreover, both a cDNA probe and polyclonal antibody derived from renal cubilin detected gastrointestinal cubilin in the same pattern as determined by lF-cbl binding activity and as previously reported for canine gastrointestinal binding and absorption of oral cobalamin (Baker et al., 1958; Drapanas et al., 1963; Fleming et al., 1962; Marcoullis and Rothenberg, 1981). Conversely, antibody raised against purified canine ileal cubilin demonstrated cross immunoreactivity with canine renal cubilin in a previous study (Fyfe et al., 1991). However, recent reports of a null mutation in the cubilin gene of a human individual exhibiting both intestinal IF-cbl malabsorption and urinary loss of cubilin ligands provide the most elegant and conclusive demonstration that ileal and renal cubilin functions are expressions of the same gene (Aminoff et al., 1999; Kozyraki et al., 1999). The findings reported here will aid in determining the basis of the analogous disorder in dogs. Previous immunoelectron microscopic study (Levine et al., 1984) of intracellular cubilin distribution during enterocyte maturation from crypt to villus 100 tip of ileal mucosa showed that synthesis of cubilin protein could be easily detected in the mid-villus cells. The expression level of cubilin dramatically increased in the villus tips, and expression of cubilin was extremely low in the crypt cells. Northern blot of canine cubilin mRNA expression in this study showed that cubilin transcripts could only be detected in the early cell fractions assumed to represent villus cell populations. These results were supported by the parallel ALP activity which is known to be expressed only in villus cells. However, the northern blot of crypt specific canine CRP-ductin showed a conflicting result. CRP-ductin was isolated from cDNA libraries of mouse intestinal crypt cells (Cheng et al., 1996). In situ hybridization in the same study showed that in the small intestine, CRP-ductin mRNA is expressed in crypt cells at all stages of differentiation from the stem cells to the terminally differentiating cells of the crypt top, but not in the mature cells of the villus. A recent study (Takito et al., 1999) suggested that mouse CRP-ductin, human DMBT1, rabbit hensin and ebnerin are the products of alternative splicing of a single gene. The transcript of this gene for all four family members could be detected in the small intestine, but the crypt specificity has only been determined in mouse. The conflicting results for the ALP activity and the canine CRP-ductin expression may have been due to incomplete cell fractionation. The incubation condition may have been over-extensive which caused both villus and crypt cells to fall-off in the early fractions. The crypt specificity of canine CRP-ductin also needs to be confirmed. To eliminate all these concerns, other crypt specific markers could be used to determine the origin of cell population in each fraction, 101 such as thymidine kinase. Alternatively, in situ hybridization of canine cubilin directly on the dog ileum sections could be used to determine the cubilin mRNA expression along the crypt to villus axis. 102 References Aminoff M, Carter JE, Chadwick RB, Johnson C, Grasbeck R, Abdelaal MA, Broch H, Jenner LB, Verroust PJ, Moestrup SK, Chapelle A de la, and Krahe R. (1999) Mutations in CUBN, encoding the intrinsic factor-vitamin B12 receptor, cubilin, cause hereditary megaloblastic anaemia 1. Nat Genet 21: 309-313. Baker SJ, MacKinnon NL, and Vasudevia P. (1958) The site of absorption of orally administered vitamin B12 in dogs. Ind. J. Med. Res. 46: 812-817. Baricault L, Galceran M, Ronco PM, Trugnam G, and Verroust PJ. (1995) Unusual processing of gp280, a protein associated with the intennicrovillar areas of yolk sac epithelial cells: plasma membrane delivery of immature protein. Biochem. Biophys. Res. Comm. 212: 353-359. Batt M, and Horadagoda NU. (1989) Gastric and pancreatic intrinsic factor- mediated absorption of cobalamin in the dog. Am. J. Physiol. 257 (Gastrointest. Liver Physiol. 20) G344-G349. Batuman V, Verroust PJ, Navar GL, Kaysen JH, Goda FO, Campbell WC, Simon E, Pontillon F, Lyles M, Bruno J, and Hammond TG. (1998) Myeloma light chains are ligands for cubilin (gp280). Am. J. Physiol. 275 (Renal Physiol. 44): F246- F254. Birn H, Fyfe J, Jacobsen C, Mounier F, Verroust PJ, erkov H, Willnow T, Moestrup SK, and Christensen El. (In press) Cubilin is an albumin binding protein important for renal albumin reabsorption. J. Clin. Invest. Bim H, Verroust PJ, Nexo E, Hager H, Jacobsen C, Christensen El, and Moestrup SK. ( 1997) Characterization of an epithelial ~460-kDa protein that facilitates endocytosis of intrinsic factor-vitamin B12 and binds receptor- associated protein. J. Biol. Chem. 42: 26497-26504. Cheng H, Bjerknes M and Chen H. (1996) CRP-ductin: a gene expressed in intestinal crypts and in pancreatic and hepatic ducts. Anat Rec 244: 327-343. Drapanas T, Williams JS, McDonald JC, Heyden W, Bow T, and Spencer RP. (1963) Role of the ileum in the absorption of vitamin B12 and intrinsic factor (NF). Clin. Sci. 184: 337-341. Eaton DM, Livingston JH, Seetharam B, and Puntis JWL. (1998) Overexpression of an unstable intrinsic factor-cobalamin receptor in Imerslund-Grasbeck syndrome. Gastroenterology 115: 173-176. 103 Fleming WH, King ER, Galloway RA, and Roche JJ. (1962) The site of absorption of orally administered Cow-labeled vitamin B12 in dogs: the effect of dose. Gastroenterology 42: 164-168. Fyfe JC. (1993) Feline intrinsic factor (IF) is pancreatic in origin and mediates ileal cobalamin (cbl) absorption. J. Vet. Intern. Med. 7: 133 (abstract). Fyfe JC, Kurzhals RL, Lassaline ME, Henthorn PS, Alur PRK, Wang P, Wolfe JH, Giger U, Haskins ME, Patterson DF, Sun H, Jain S, and Yuhki N. (1999) Molecular basis of feline B-glucuronidase deficiency: an animal model of mucopolysaccharidosis VII. Genomics 58: 121-128. Fyfe JC, Ramanujam KS, Ramaswamy K, Patterson DF, and Seetharam B. (1991) Defective brush-border expression of intrinsic factor-cobalamin receptor in canine inherited intestinal cobalamin malabsorption. J. Biol. Chem. 266: 4489- 4494. Guéant J-L, Chery C, Namour F, Aimone—Gastin I, and Wustinger M (Reply, Seetharam B). (1999) Decreased affinity of urinary intrinsic factor-cobalamin receptor in a case of Grasbeck-Imerslund syndrome. Gastroenterology116: 1274-1276. Hagedorn CH and Alpers DH. (1977) Distribution of intrinsic factor-vitamin B12 receptors in human intestine. Gastroenterology 73: 1019-1022. Hammad SM, Stefansson S, Twal WO, Drake CJ, Fleming P, Remaley A, Brewer HB, and Argraves WS. (1999) Cubilin, the endocytic receptor for intrinsic factor- vitamin B12 complex, mediates high-density lipoprotein holoparticle endocytosis. Proc. Natl. Acad. Sci. USA 96: 10158-10163. Hammond TG, Verroust PJ, Majewski RR, Muse KE, and Oberley TD. (1994) Heavy endosomes isolated from the rat renal cortex show attributes of intennicrovillar clefts. Am. J. Physiol. 267 (Renal Fluid Electrolyte Physiol. 36: F516-F527. Kahn BB. (1992) Facilitative glucose transporters: regulatory mechanisms and dysregulation in diabetes. J. Clin. Invest. 89: 1367-1374. Kapadia CR. (1995) Vitamin B12 in health and disease: part l--inherited disorders of function, absorption and transport. Gastroenterologist 3: 329-344. Kessler M, Acuto O, Storelli C, Murer H, Miiller M, and Semenza G. (1978) A modified procedure for the preparation of efficiently transporting vesicles from small intestinal brush border membranes. Biochim. Biophys. Acta 506: 136-154. 104 Kozyraki R, Fyfe J, Kristiansen M, Gerdes C, Jacobsen C, Cui S, Christensen El, Aminoff M, Chapelle A de la, Krahe R, Verroust PJ, and Moestrup SK. (1999) The intrinsic factor-vitamin B12 receptor, cubilin, is a novel high affinity apolipoprotein A-I receptor facilitating endocytosis of high-density Iipoprotein. Nat. Med. 6: 656-661. Kozyraki R, Kristiansen M, Silahtaroglu A, Hansen C, Jacobsen C, Tommerup N, Verroust PJ, and Moestrup SK. (1998) The human intrinsic factor-vitamin B12 receptor, cubilin: Molecular characterization and chromosomal mapping of the gene to 10p within the autosomal recessive megaloblastic anemia (MGA 1) region. Blood 91: 3593-3600. Kristiansen M, Kozyraki R, Jacobsen C, Nexo E, Verroust PJ, and Moestrup SK. (1999) Molecular dissection of the intrinsic factor-vitamin B12 receptor, cubilin, discloses regions important for membrane association and ligand binding. J. Biol. Chem. 274: 20540-20544. Le Panse S, Ayani E, Nielsen S, Ronco P, Verroust P, and Christensen El. (1997) Internalization and recycling of glycoprotein 280 in epithelial cells of yolk sac. Europ. J. of Cell Biol. 72: 257-267. Levine JS, Allen RH, Alpers DH, and Seetharam B. (1984) lmmunocytochemical localization of the intrinsic factor-cobalamin receptor in dog-ileum: distribution of intracellular receptor during cell maturation. J. Cell Biol. 98: 111 1-1118. Lindblom A, Quadt N, Marsh T, Aeschlimann D, Mergelin M, Mann K, Maurer P, and Paulsson M. (1999) The intrinsic factor-vitamin B12 receptor, cubilin, is assembled into trimers via a coiled-coil a-helix. J. Biol. Chem. 274: 6374-6380. Lowry OH, Rosebrough NJ, Farr AL, and Randall RJ. (1951) Protein measurement with the folin phenol reagent. J. Biol. Chem. 193: 265-275. Marcoullis G and Rothenberg SP. (1981) Intrinsic factor-mediated intestinal absorption of cobalamin in the dog. Am. J. Physiol. 241 (Gastrointest. Liver Physiol. 4) GZ94-G299. Moestrup SK, Kozyraki R, Kristiansen M, Kaysen JH, Rasmussen HH, Brault D, Pontillon F, Goda FO, Christensen El, Hammond TG, and Verroust PJ. (1998) The intrinsic factor-vitamin B12 receptor and target of teratogenic antibodies is a megalin-binding peripheral membrane protein with homology to developmental proteins. J. Biol. Chem. 273: 5235-5242. Sambrook J, Fritch EF, and Maniatis J. (1989) Molecular Cloning: A Laboratory Manual, 2nd ed. Cold Spring Harbor, NY, Cold Spring Harbor Laboratory Press. 105 Seetharam B, Alpers DH, and Allen RH. Isolation and characterization of the ileal receptor for intrinsic factor-cobalamin. J. Biol. Chem. 256: 3785-3790. Seetharam B, Bagur SS, and Alpers DH. (1981) Interaction of receptor for intrinsic factor-cobalamin complex with synthetic and brush-border lipids. J. Biol. Chem. 256: 9813-9815. Seetharam B, Bagur SS, and Alpers DH. (1982) Isolation and characterization of proteolytically derived ileal receptor for intrinsic factor-cobalamin. J. Biol. Chem. 257: 183-189. Seetharam B, Bakke JE, and Alpers DH. (1983) Binding of intrinsic factor to ileal brush border membrane in the rat. Biochem. Biophys. Res. Comm. 115: 238- 244. Seetharam B, Christensen El, Moestrup SK, Hammond TG, and Verroust PJ. (1997) Identification of rat yolk sac target protein of teratogenic antibodies, gp280, as intrinsic factor-cobalamin receptor. J. Clin. Invest. 99: 2317-2322. Simpson KW, Alpers DH, Wille J De, Swanson P, Farmer S, and Sherding RG. (1993) Cellular localization and hormonal regulation of pancreati S c intrinsic factor secretion in dogs. Am. J. Physiol. 265 :(Gastrointest. Liver Physiol. 28) G178-G188. Takito J, Yan L, Ma J, Hikita C, Vijayakumar S, Warburton D, and Al-Awqati Q. (1999) Hensin, the polarity reversal protein, is encoded by DMBT1, a gene frequently deleted in malignant gliomas. Am J Physiol 277 (Renal Physiol 46): F277-F289. Traber PG, Yu L, Wu GD, and Judge TA. (1992) Sucrase-isomaltase gene expression along crypt-villus axis of human small intestine is regulated at level of mRNA abundance. Am J Physiol 262 (Gastrointest Liver Physiol 25): G123- G130. Weiser MM. (1973) Intestinal epithelial cell surface membrane glycoprotein synthesis. 1. An indicator of cellular differentiation. J.Biol. Chem. 248: 2536-2541. Xu D, Kozyraki R, Newman TC, and Fyfe JC. (1999) Genetic evidence of an accessory activity required specifically for cubilin brush-border expression and intrinsic factor-cobalamin absorption. Blood 94: 3604-3606. 106 CHAPTER 4 LINKAGE ANALYSIS OF CANINE SELECTIVE COBALAMIN MALABSORPTION WITH CUBN AND MEGALIN LOCI 107 Introduction Hereditary megaloblastic anemia 1, (MGA 1, OMIM #261100), also known as Imerslund-Grasbeck syndrome (lG-S), is a rare autosomal recessive disorder characterized by selective intestinal cobalamin (vitamin B12) malabsorption leading to juvenile-onset severe megaloblastic anemia and/or neurologic manifestations. Most patients also exhibit mild proteinuria. The disorder has been described in families in Nonivay (Imerslund 1960), Finland (Grasbeck et al., 1960), Israel (Ben-Bassat et al., 1969), Saudi Arabia (Abdelaal and Ahmed, 1991), Turkey (Altay et al., 1995; Altay and Cetin, 1999) and sporadically worldwide. MGA 1 was mapped to chromosome 10p12.1 in Finnish, Norwegian and a Saudi Arabian kindred (Aminoff et al., 1995, 1999). One candidate gene for lG-S was cubilin, a 460 kDa protein found in the apical membrane domain of epithelial cells of the distal small intestine, and renal proximal tubules (Seetharam et al., 1997; Birn et al., 1997). Human (GenBank accession # AF034611; Kozyraki et al., 1998) and rat (GenBank accession #AF022247; Moestrup et al., 1998) cubilin were recently cloned. The human CUBN locus was mapped within the MGA 1 locus by fluorescence in situ hybridization, radiation hybrid mapping, and screening of YAC clones (Moestrup et al., 1998). Two mutations, a missense mutation in CUB domain 8 and an intronic point mutation activating a cryptic splice site within CUB domain 6, were recently demonstrated in Finnish kindreds (Aminoff et al., 1999). A canine model with selective intestinal Cbl malabsorption was identified with clinical and laboratory features similar to those seen in human (Fyfe et al., 108 1991a). Immunoelectron microscopy and cell fractionation studies showed failure of cubilin expression on the apical brush-border membrane of either tissue (Fyfe et al., 1991b). Characterization of the urine proteins of affected dogs showed ApoA-1 (Kozyraki et al., 1999), albumin (Birn et al., in press), haptoglobin, lgG light chains and other unidentified proteins (Fyfe et al., unpublished data), which are the same as the urine proteins identified in the patient with a cryptic splice site (null) mutation of cubilin. To further determine whether defective function of canine cubilin is caused by a mutation within its coding sequence, which is the most common reason for a protein trafficiking disorders, we perfomed the linkage analysis to determine whether the cubilin gene locus and the disease locus are genetically linked. The predicted protein structure of human and rat cubilin showed no sequence compatible with a transmembrane domain nor a signal for glycosylphospho-tidylinositol anchor addition. Parallel with the predicted structure, the protein showed only a weak association with the plasma membrane (Moestrup et al., 1998). All these data suggested that cubilin is a peripheral membrane protein. As a peripheral membrane protein without internalization signals, cubilin-mediated endocytosis of the lF-Cbl complex may be facilitated by another membrane protein. Megalin, a transmembrane protein with an endocytic signal sequences in the cytoplasmic domain, interacts with cubilin in vitro (Moeatrup et al., 1998), colocalizes with cubilin in vivo (Sahali et al., 1988, 1993, Birn et al., 1997, Moestrup et al., 1998) and exhibits coregulation of expression (hammad et al., 2000). Megalin, therefore, has been suggested to 109 play an important role in facilitating the endocytosis of the IF-Cbl complex. Therefore, a defect in the interaction between cubilin and megalin may be another reason for failure of cubilin expression on apical brush border membranes. To determine whether the defective interaction between megalin and cubilin caused the Cbl malabsorption in this canine model, we also conducted linkage analysis between canine megalin and selective intestinal cobalamin malabsorption in this dog family. 110 Materials and Methods Cubilin linkage study Dogs in the linkage studies were members of a breeding colony maintained at Michigan State University; protocols for husbandry and tissue collection were approved by the MSU All-University Committee for Animal Use and Care. The parents and 23 offspring, including 13 affected and 10 clinically- normal carriers, of matings between an affected female and an obligate carrier male were studied. The disease phenotype of each dog was determined by the following criteria: affected dogs became inappetent and failed to gain weight after 10-14 weeks of age; they had low serum cobalamin concentrations, methylmalonic aciduria, mild nonregenerative anemia, and neutropenia. And all hemotologic, metabolic, and growth abnormalities were reversed by parenteral cobalamin administration. Due to the carrier X affected matings strategy, offspring determined as not affected were considered obligate carriers of selective cobalamin malabsorption. Genomic DNA of normal dogs was isolated from frozen liver samples. Approximately 0.5 g of frozen tissue from each dog was ground into fine power in liquid nitrogen and gently spread onto extraction buffer (10 mM Tris-HCI, pH 8.0, 0.1 M EDTA, 20 pglml pancreatic RNAase, 0.5% SDS), allowing the powder to become submerged. Tissue suspensions were incubated with 100 pglml proteinase K at 55°C overnight and were extracted in 0.5 M Tris-HCI (pH 8.0) equilibrated phenol, phenol and chloroform mixture, and chloroform sequentially and precipitated by 10 M ammonium acetate (0.2 vol/vol) and 100% ethanol (2 111 vol/vol). Isolated DNA was washed in 70% ethanol and resuspended in TE pH 8.0, and quantified by spectrophotometry. Genomic DNA was amplified by PCR using various combinations of the primers 838F, 5’-AGCCTGCGTGCTGGACATCGAC-S’; 947F, 5’- GGATGGCAAGGAAATGGATATAGT—3’; 1150R, 5’-TGGGTGGCAGCCTC- CATTAT'I'GA-3’; and 1341 R 5’-CCAGCCCAACCTGATTCACACTTA-B’ designed from the canine cubilin cDNA sequence. 50 pL reaction mixtures contained 25 mM TAPS (pH 9.3), 50 mM KCI, 2 mM MgClz, 1 mM 2-mercaptoethanol, 0.4 mM each deoxynucleotide, 0.5 pM each primer, 500 ng of DNA template, and 2.5 U of TaKaRa LA Taq polymerase (PanVera, Corp., Madison, WI). After a hotstart denaturation at 94°C for 2 min, reactions were carried out for 35 cycles of denaturation at 98°C for 20 s and annealing / extension at 68°C for 20 min. Reaction products were gel purified and partially sequenced using the above PCR primers. According to the sequencing results, a 0.9 kb intron of both an obligate carrier male and an affected female were amplified using primer 947F and 1150R. The PCR products were gel purified and cloned into the plasmid PCRTM ll (Invitrogen®, San Diego, CA) at 16°C overnight. Recombinant plasmids were transformed into DH5 a competent cells. Six individual clones from each dog were purified by QIAGEN plasmid purification kit and sequenced to identify intronic sequence variations. DNA templates used for PCR amplification of a cubilin intron variation (CIV) were isolated from snap frozen liver or from fresh blood samples. The DNA 112 from frozen tissues was isolated by the same protocol as previously described. The DNA from whole blood cells was isolated by the following procedure. The whole blood cells were Iysed by ST buffer (0.32 M sucrose 10 mM Tris, pH7.6, 5 mM MgCl2 and 1% Triton X-100) and the leukocyte nuclei were resuspended in NE buffer (75mM NaCl and 25 mM EDTA pH 8.0) and 0.1% SDS. After sequential incubation with proteinase K and RNAse A, the genomic DNA was extracted and precipitated as from frozen tissue described previously. To PCR amplify the intron variation, the primers (CIVF, 5’-GATCACAGGCCTACAGCTC- CATT-3’ and CIVR, 5’-CCAGGCCAACCAGAGATCTTCTA-3’) were designed to flank a 17 base insertion sequence. PCR produced allele-specific amplification products of 199 and 182 bp. PCR reactions were 50 pL, containing 50 mM KCI, 10 mM Tris-Cl (pH 8.3), 2 mM MgClz, 0.37 mM each deoxynucleotide, 0.25 pM each primer, and 100 ng of DNA template. Taq DNA polymerase (2.5 U) was added after a 5 min hotstart at 95°C, and PCR reactions were carried out for 28 cycles of denaturation for 30 s at 95°C, annealing for 60 s at 65°C, and extension for 120 s at 72°C. The reaction products were electrophoresed in TAE buffer on 4% agarose gels and visualized by ethidium bromide staining. Southern blotting was performed on genomic DNA of carrier and affected dogs isolated from frozen liver as described above. Genomic DNA was digested by Barn HI, EcoRl, Hind Ill, Pst l, and Xba I and 5 pg of each digested DNA was electrophoresed on a 1% agarose gel. DNA was transferred to a nylon membrane, and the blot was hybridized by standard methods (Sambrook et al., 1989) to a canine cubilin cDNA probe (bp 947-1150). 113 Megalin linkage study: The primers used for canine megalin DNA amplification were designed from a conserved region of human and rat megalin and human low-density lipoprotein related protein (LRP1). The primer: Meg fonivard, 5’-GGCTCCT- ACATCTGTAAGTGTGC-3’; Meg reverse, 5’-CACAGACTTG‘ITGGTI’CCATCC- 3’ were designed from human LRP1 exon 56 and exon 60 respectively. Reverse transcribed polymerase chain reaction (RT-PCR) was performed using above primers to amplify canine megalin cDNA from total RNA of dog kidney which was isolated from snap frozen dog kidney tissue using Trizol® reagent (GibCo protocol). The PCR product was gel purified and sequenced using the same pair of primers. The same pair of primers was used to amplify the canine genomic megalin fragment. The 50 pl reaction mixture contained 300 mM Tris-HCI (pH 8.5), 75 mM ammonium sulfate, 2.5 mM MgClz, 0.25 mM each deoxynucleotide, 0.5 pM each primer, and 300 ng of dog genomic DNA template. 2.5 U of AmpliTaq polymerase (Perkin Elmer, Foster city, CA) was added after a hotstart denaturation at 95°C for 5 min, then reactions were carried out for 32 cycles with 30s denaturation at 95°C, 305 annealing at 58°C and 2 min extension at 72°C. The single PCR product from an obligate carrier dog was gel purified and sequenced. In order to separate two PCR products (both are around 2.1 kb) from an affected dog, a new fonivard primer, 1501F, 5’- CATTGCCTCTCTGTCTCTGACCT-3’, was designed from the middle exon and 114 used for PCR amplification under the same reaction conditions, but without the hotstart, to produce smaller PCR products (about 700 bp). The two smaller PCR products of the affected dog were gel purified and cloned individually in the plasmid pCRTM ll (Invitrogen, San Diego, CA). Two purified plasmids containing each PCR product were sequenced separately. The primers, 1553F, 5’-GAGCACTGTGTGGATGTCAA-3’; 2009R, 5’- CATGGAGAGTCCAGTATAGG-3’ were designed to flank an intronic insertion of 68 bp defined from canine megalin sequence. PCR amplification of this canine megalin intron variation was performed on DNA isolated from frozen liver or fresh blood samples using the same methods as above. Totally 24 dog samples with confirmed phenotype were used for this study, including one normal dog, 9 carrier dogs and 14 affected dogs for selective cobalamin malabsorption. The conditions for the 50 pl PCR reaction were as same as for the canine megalin genomic DNA amplification except using 1 min extension time instead of 2 min. Allele specific PCR products were separated on a 4% agarose gel in 1X TAE buffer and visualized by ethidium bromide staining. 115 Results The intron/exon structure of the canine CUBN was defined in the region of EGF domains 4-7 by amplification of genomic DNA by PCR, using primer sequences from the cDNA (Figure 4.1, panel A). Sequencing of the PCR products demonstrated concensus splice donor and acceptor sequences at the points where the genomic sequence diverged from the cDNA sequence. In the 5’ to 3’ direction, 4 introns (2.2, 0.9, 2.0 and 0.2 kb) and 3 included exons (cDNA sequences 948-1079, 1080-1175, and 1176-1294; Gly292-Pro335, Gly336- Leu367, and Gly368-Ile407) were defined. The intron/exon boundaries did not correspond to the protein structural boundaries of the EGF domains. A canine CUBN variation was determined within the 0.9 kb intron between nucleotides 1079 and 1080 of the cDNA sequence in EGF domain 5 (Panel A). The intron between PCR primers 947F and 1150 R of a heterozygous male dog (F274 Figure 4.1) was amplified, and the product was cloned. Sequencing of six individual clones demonstrated a 17 bp sequence (5’-CAGAACA'I‘I'G'I'T'I'ATGC- 3’) in three of the clones inserted 187 bp 5’ of the 3’ intron/exon boundary. Sequencing of 6 clones of the intron similarly amplified from a homozygous female (F284, Figure 4.1) demonstrated that she was likely (P>0.98) homozygous for the allele containing the 17 bp insert. Single hybridizing bands of 5-10 kb, identical in carrier and affected dog DNA, were observed on Southern blots of canine genomic DNA digested with 6 different restriction enzymes and hybridized to a probe of CUBN cDNA sequence between bp 947 and 1150, thus 116 838F 947.: - ’ 2.2 kb 0.9 2.0 kb I ’ ‘ ‘ a,” 1150a ‘~‘ 1341a ’ ’ ~ ‘ e ’ ’ crvr= ~ 2 fi 61" AG I J " B 17 bp variation C'VR F274 F284 * * ‘k * 1k Figure 4.1: Independent segregation of canine I-GS and CUBN loci. (A) Four introns (horizontal lines) and included exons (vertical boxes) were defined by PCR amplification using CUBN cDNA primers and sequencing the products. A 17—bp variation (vertical line) was found in the 0.9-kb intron for which dog F274, an obligate carrier of canine l-GS, was heterozygous and dog F284, an affected dog, was homozygous. (B) Solid symbols indicate l-GS affected dogs, half-solid symbols indicate obligate carriers, squares are males, and circles are females. DNA from offspring of matings between dogs F274 and F284 was amplified by PCR using primers flanking the 17-bp variation, producing allele- specific products of 199 and 182 bp. Results of 10 offspring are shown, with the stars indicating recombinants. 117 BamHl EcoRl Hind Ill Pstl Xbal .C A . C} A C. ’7 Wind Ill 2 Figure 4.2: lntragenic cubilin marker is located in a unique region of the canine genome.Southern blots of canine genomic DNA digested with 6 bp recognition-site restriction endonucleases were prepared and hybridized to a canine cubilin cDNA probe flanking the intron containing an identified variation (see figure 4.1). Single hybridizing bands of 5—10 kb, identical in carrier and affected dog DNA were observed, thus confirming that the identified 17-bp marker variation was in a unique region of the canine genome. C: carrier dog, A: affected dog. 118 confirming that the identified 17-bp variation was in a unique region of the canine genome (Figure 4.2). A CUBN allele-specific PCR test was designed to examine the 17 bp insertion variation in affected and carrier offspring of affected X carrier matings (phase-known double backcross matings) performed between dogs F284 and F274 (Figure 4.1, Panel B). The affected offspring from these matings are homozygous and clinically normal Iittermates which are heterozygous at the disease locus. The PCR test confirmed that the clinically affected dam was homozygous and the clinically normal, obligate carrier sire was heterozygous for the 17 bp insert. However, 13 recombinants were detected among 23 offspring of these matings, a recombination fraction (0.56) that did not differ from the recombination fraction (0.5) expected under the hypothesis of independent segregation of the CUBN and disease loci (X2=0.39, df=1, P=0.53). To test another candidate gene for this canine model with selective intestinal cobalamin malabsorption, a 2290 bp-canine megalin genomic DNA fragment was amplified by PCR using primer pairs designed from conserve regions of human and rat megalin. The alignment between dog megalin cDNA amplified from dog kidney total RNA by RT—PCR using the same pair of primers and this canine genomic DNA fragment defined the partial intron/exon structure of canine megalin. This region contained two introns (1.1 kb and 0.6 kb) and 1 included exon (156 bp) (Figure 4.3, Panel A). The canine megalin cDNA fregment showed 88% identity to human megalin, which confirmed the sequence specificity. 119 .3000. 000508000: or 00265 octamto cm :0 0:300: 0:... .0: 03 0:0 000 :0 $0302: 050000.220 050309: .:o=0:0> 00-00 0:: 0:2:0: 0.0:..5 050: mun. >0 005.9:0 003 9.50:0 E0: <20 00.080: 00 00.2.0 0:0 .00.0E 0.0 00035 .000 .950: 0 00:00.05 .onE>m->EEO 05 0:0 .90.:00 00.09.00 0020:. 0.00.50 0:00.20: .0000 00000.00 m0. 0:00.05 0.88% 0_.ow Am. .0.0>.0:0 0000.5. :0.— .0xL0E .0E0E. 00 000: 002, 0:0 :22. 9.0.0 05 :_ 00:050. 002, 080 0:00 :o.~0.:0> 3.00 < 20:00:: 05 050:0:000 0:0 0:060: 0020050 5.000.: :0: 0:0 :0E2. E0: 00:0.000 0.0800 (200 5.000;: 0.50: 50005380 mun. .5 005000 0:03 068 :90 00020:. 0:0 .005. .0.:oN_:o£ 0:055 02:. 2. ._00_ 5.80:. 0:0 m0. 05:00 no 5000.500 E00:0:00:. ”0.0 0.50.“. 120 .4. a. E Home I \ , It \ LIIIJ MT 8. m... rflT e. E i m 52: u_ 82 u. .55 121 A canine megalin intron variation was determined within the 0.5 kb intron. Subclone and sequence results of the PCR amplified 0.5 kb intron of an affected dog genomic DNA showed that one allele contained a 68 bp insertion (AGCTGGTCTGTITTCTT(T28)TGT(T15)TT), but no insertion was found in the other allele. This result was in conflict with the disease locus, which is homozygous for the affected dogs. In contrast, the obligate carrier dog, which was heterozygous for the disease locus, showed homozygousity for the megalin intron variation, with no insertion in either allele. At this point, the data suggested that the disease locus was not genetically linked with the megalin locus in this family of dogs with selective intestinal Cbl malabsorption. Further family linkage study with 20 offspring (12 affected dogs and 8 carrier dogs) (Figure 4.3), using allele specific PCR amplification with primer 1553F and 2009R, producing a 390 bp product for the normal allele and a 456 bp product for the insertion allele, showed 10 recombinants (0.5 recombination fraction) and confirmed independent segregation of the disease locus and the megalin locus in this family of dogs with selective intestinal Cbl malabsorption. 122 Discussion Cubilin is a biologically compelling candidate gene for selective intestinal cobalamin malabsorption with proteinuria. Cubilin is a multi-ligand receptor expressed in the small intestinal epithelium where it mediates absorption of cobalamin via binding of lF-Cbl complexes (Seetharam et al. 1997) Cubilin is also expressed in the renal proximal tubule epithelium where it mediates reabsorption of specific proteins filtered by the glomerulus (Batuman et al., 1998; Birn et al., 2000; Fyfe et al., unpublished data). CUBN mutations causing selective intestinal cobalamin malabsorption with proteinuria in Finnish kindreds have been defined (Aminoff et al., 1995; Kozyraki et al., 1998). The canine model of selective intestinal cobalain malabsorption with proteinuria is a well-defined, simple autosomal recessive disorder for which CUBN was also an attractive candidate gene. Intrinsic factor and transcobalamin- ll functions of the affected dogs were demonstrated to be normal, and immunoelectron microscopy revealed lack of cubilin in the apical membrane of distal intestinal epithelial cells (Fyfe et al., 1991a). Measurements of cubilin ligand-binding activity and western blotting of fractionated cells demonstrated that the cubilin protein was expressed in affected dog ileal and proximal tubule epithelial cells but did not reach the apical brush border membrane. Cubilin purified by detergent solubilization and affinity chromatography from normal and affected dog renal cortex comigrated on nonreducing SDS-PAGE gels but yielded different peptides upon proteolytic digestion. Additionally, Asn-linked oligosaccharides on affected dog cubilin were in an Endo H sensitive form, 123 suggesting retention in the endoplasmic reticulum (ER) and failure of the nascent protein to make its way through the biosynthetic pathway (Fyfe et al., 1991b). These findings were analogous to findings in a number of disorders of secretory and membrane protein expression in which mutations of specific genes cause failure of the translation product to fold properly and to remain associated with ER chaperone proteins, to self-aggregate, or to be degraded rapidly (Kopito 1999; Bross et al., 1998; Kim and Arvan 1998). With the exception of abetalipoproteinemia (MTP, MIM #200100), all inherited ER retention/degradation disorders described to date are attributed to mutations in the coding sequence of the particular secreted or membrane protein. The similarities of the cloned canine CUBN cDNA sequence with those of other species and the match of cubilin peptides to the deduced amino acid sequence of the canine clone confirmed its identity (Chapter 2). Elimination of linkage of an lntragenic CUBN marker to the inherited disorder in this canine family indicates that failure of apical membrane expression of cubilin and the resultant selective cobalamin malabsorption is caused by dysfunction of a gene product whose locus maps 2 50 cM from CUBN in the canine genome. Because cellular physiology of humans and dogs is highly homologous, these findings suggest that human l-GS may exhibit locus heterogeneity in addition to allelic heterogeneity at the CUBN locus. Human l-GS remains to be mapped in kindreds of various ethnic backgrounds. Genes unlinked to the human MGA 1 locus which may explain our findings include the gene encoding the endocytic receptor, megalin (gp330, LRP2), 124 mapped to 2q31 (Chowdhary et al. 1995) which has been demonstrated to associate and co-localize with cubilin (Birn et al. 1997; Moestrup et al. 1998). It has been postulated that megalin may anchor cubilin to the apical membrane and mediate its vesicular trafficking because cubilin lacks a transmembrane domain or other obvious membrane anchoring signal. Megalin, which is closely related to LRP also binds the urokinase-inhibitor complex (Nykjaer et al., 1997; Herz et al., 1992), and may perform a similar function in regard to both the urokinase receptor and cubilin-ligand complexes. The rejection of linkage between the canine megalin locus and the disease locus of this canine model with selective cobalamin malabsorption indicates that the failure of membrane expression of cubilin in affected dogs was caused by a defect of a gene other than megalin. To locate the disease-causing gene for this canine model with selective cobalamin malabsorption, instead of genome-wide screening, linkage analysis of candidate genes were performed. Another candidate gene product is an ER chaperone, Iipoprotein related protein-associated protein 1 (LRPAP 1) or RAP. RAP interacts with cubilin in vitro and maps to 4p16.3 (VanLeuven et al. 1995) of humans. By analogy to the function of association with megalin (LRP2) and the low density lipoprotein receptor-related protein 1/02-macroglobulin receptor (LRP1), RAP may mediate proper folding of cubilin and prevent premature ligand binding to cubilin within the ER and later compartments of the biosynthetic pathway (Bu and Schwartz 1998). RAP is essential for the proper folding and disulfide bond formation of LRP1, findings supported by ER retention 125 and rapid degradation of LRP1 and other members of the LDL-receptor gene family in RAP-knockout mice (Willnow TE et al., 1996; Oberrnoeller et al., 1998). Future investigations of canine selective intestinal cobalamin malabsorption in this family will focus initially on the LRPAP1 or RAP locus. 126 References Abdelaal MA and Ahmed AF. (1991) Case report: Imerslund-Grasbeck syndrome in a Saudi family. Acta Padiatr Scand 80: 1109-1112. Altay C, Cetin M, Giimri’ik F, Irken G, Yetgin S. and Laleli Y. (1995) Familial selective vitamin B12 malabsorption (Imerslund-Grasbeck syndrome) in a pool of Turkish patients. Pediat Hematol Oncol 12:19-28. Aminoff M, Tahvanainen E, Grasbeck R, Weissenbach J, Broch H, and de la Chapelle A. (1995) Selective intestinal malabsorption of vitamin B12 displays recessive mendelian inheritance: assignment of a locus to chromosome 10 by linkage. Am J Hum Genet 57: 824-31. Aminoff M, Carter JE, Chadwick RB, Johnson C, Grasbeck R, Abdelaal MA, Broch H, Jenner LB, Verroust PJ, Moestrup SK, Chapelle A, and Krahe R. (1999) Mutations in CUBN, encoding the intrinsic factor-vitamin B12 receptor, cubilin, cause hereditary megaloblastic anaemia 1. Nat Genet 21: 309-313. Batuman V, Verroust PJ, Navar GL, Kaysen JH, Goda FO, Campbell WC, Simon E, Pontillon F, Lyles M, Bruno J, and Hammond TG. (1998) Myeloma light chains are ligands for cubilin (gp280) Am J Physiol. 275(R44): F246-F254. Ben-Bassat I, Feinstein A. and Ramot B. (1969) Selective vitamin B12 malabsorption with proteinuria in lsraeI-—Clinical and genetic aspects. Israel J Med Sci 5: 62-69. Birn H, Verroust PJ, Nexo E, Hager H, Jacobsen C, Christensen El, and Moestrup S. (1997) Characterization of an epithelial ~460-kDa protein that facilitates endocytosis of intrinsic factor-vitamin B12 and binds receptor- associated protein. J Biol Chem 272(42): 26497-26504. Birn H, Fyfe JC, Jacobsen C, Mounier F, Veroust PJ, Qrskov H, Willnow T, MoestrupSK, and Christensen El. (2000) Cubilin is an albumin binding protein important for renal albumin reabsorption. J Clin Invest (in press). Bork P and Beckmann G. (1993) The CUB domain. A widespread module in developmentally regulated proteins. J Mol Biol 231: 539-545. Bross P, Andresen BS, and Gregersen N. (1998) Impaired folding and subunit assembly as disease mechanism: the example of medium-chain acyl-CoA dehydrogenase deficiency. Prog Nucleic Acid Res Mol Biol 58: 301-337. Bu G and Schwartz AI (1998) RAP, a novel type of ER chaperone. Trends Cell Biol 8: 272-276. 127 Chowdhary BP, Lundgren S, Johansson M, Hjalm G, Akerstrom G, Gustavsson I, and Rask L. (1995) In situ hybridization mapping of a 500-kDa calcium-sensing protein gene (LRP2) to human chromosome region 2q31->q32.1 and porcine chromosome region 15q22—>q24. Cytogenet Cell Genet 71: 120-123. Fyfe JC, Giger U, Hall CA, Jezyk PF, Klumpp SA, Levine JS, and Patterson DF. (1991a) Inherited selective intestinal cobalamin malabsorption and cobalamin deficiency in dogs. Pediatr Res 29: 24-31. Fyfe JC, Ramanujam KS, Ramaswamy K, Patterson DF, and Seetharam B. (1991b) Defective brush-border expression of intrinsic factor-cobalamin receptor in canine inherited intestinal cobalamin malabsorption. J Biol Chem 266: 4489- 4494. Grasbeck R, Gordin R, Kantero I, And Kuhlback B. (1960) Selective vitamin B12 malabsorption and proteinuria in young people. Acta Med Scand 167:289-296. Herz J, Clouthier DE, and Hammer RE. (1992) LDL receptor-related protein internalizes and degrades uPA-PAl-1 complexes and is essential for embryo implantation. Cell 71 : 411-421. Imerslund O. (1960) Idiopathic chromic megaloblastic anemia in children. Acta Paediat Scand Suppl 49: 1-115. Kim PS and Arvan P (1998) Endocrinopathies in the family of endoplasmic reticulum (ER) storage diseases: disorders of protein trafficking and the role of ER molecular chaperones. Endocr Rev 19: 173-202. Kopito RR (1999) Biosynthesis and degradation of CFTR. Physiol Rev 79: S167- S173. Kozyraki R, Kristiansen M, Silahtaroglu A, Hansen C, Jacobsen C, Tommerup N, Verroust PJ, and Moesth SK. (1998) The human intrinsic factor-vitami Biz receptor, cubilin: molecular characterization and chromosomal mapping of the gene to 10p within the autosomal recessive megaloblastic anemia (MGA 1) region. Blood 91 (10): 3593-3600. Moestrup SK, Kozyraki R, Kristiansen M, Kaysen JH, Rasmussen HH, Brault D, Pontillon F, Goda F, Christensen El, Hammond TG, and Verroust PJ. (1998) The intrinsic factor-vitamin B12 receptor and targek of teratogenic antibodies is a megalin-binding peripheral membrane protein with homology to developmental proteins. J Biol Chem 273(9): 5235-5242. Nykjaer A, Conese M, Christensen El, Olson D, Cremona O, Gliemann J, and Blasi F. (1997) Recycling of the urokinase receptor upon internalization of the uPAzserpin complexes. EMBO J 16: 2610-2620. 128 Obermoeller LM, Chen Z, Schwartz AL, and Bu G. (1998) Caz+ and receptor- associated protein are independently required for proper folding and disulfide bound formation of the low density Iipoprotein receptor-related protein. J Biol Chem 273: 22374-22381. Sahali D, Mulliez N, Chatelet F, Laurent-Winter C, Citadelle D, Sabourin JC, Roux C, Ronco P, and Verroust P. (1993) Comparative immunochemistry and ontogeny of two closely related coated pit proteins. The 280-kd target of teratogenic antibodies and the 330-kd target of nephritogenic antibodies. Am J Pathol142: 1654-1667. Sahali D, Mulliez N, Chatelet F, Dupuis R, Ronco P, and Verroust P. (1988) Characterization of a 280-kD protein restricted to the coated pits of the renal brush border and the epithelial cells of the yolk sac. Teratogenic effect of the specific monoclonal antibodies. J Exp Med 167: 213-218. Sambrook J, Fritch EF, and Maniatis J. (1989) Molecular cloning: A laboratory manual, 2"d ed. Cold Spring Harbor, NY, Cold Spring Laboratory Press, 1989. Seetharam B, Christensen El, Moestrup SK, Hammond TG, and Verroust PJ. (1997) Identification of rat yolk sac target protein of teratogenic antibodies, gp280, intrinsic factor-cobalamin receptor. J Clin Invest 99(10): 2317-2322 Van Leuven F, Hilliker C, Serneels L, Umans L, Overbergh L, De Strooper B, Fryns JP, and Van den Berghe H. Cloning, characterization, and chromosomal localization to 4p16 of the human gene (LRPAP1) coding for the alpha 2- macroglobulin receptor-associated protein and structural comparison with the murine gene coding for the 44-kDa heparin-binding protein. Genomics 25: 492- 500. V\fillnow TE, Armstrong SA, Hammer RE, and Herz J. (1995) Functional expression of low density lipoprotein receptor-related protein is controlled by receptor-associated protein in vivo. Proc Natl Acad Sci USA 92: 4537-4541. 129 Future Direction The major focus of future projects will be to identify the molecular mechanism of this canine model of selective intestinal cobalamin malabsorption which closely resembles the human Imerslund-Grasbeck syndrome. In this thesis, we used linkage analysis of the disease locus with cubilin and megalin gene loci and rejected the hypothesis that cubilin or megalin was the disease causing gene product. The specific goal for future projects will be to generate new candidate genes. From this point, two approaches can be used to identify new candidate genes, which are functional analysis and positional mapping. The rejection of genetic linkage of canine l-GS with canine cubilin and megalin gene suggested that a gene product other than megalin may physically interact with cubilin and be required in the biosynthesis or apical delivery of cubilin to the plasma membrane. Identification of proteins which interact cubilin will generate functional candidate genes. Receptor associate protein (RAP), which interacts with cubilin in vitro (Moustrup et al., 1999) will be studied first. RAP is an ER chaperone-like protein (Bu et al., 1995) and may mediate proper folding of cubilin and prevent premature ligand binding to cubilin within the ER. Human and rat RAP genes are cloned and the intron/exon structure of each were identified (van Leuven et al., 1995; ). An lntragenic marker within the canine RAP gene can be identified by using the same approach as for megalin. Genetic linkage will be used to test linkage to the disease condition. 130 If the RAP gene is not linked to the disease locus, other methods, like co- immunoprecipitation, affinity chromatography, and 2-D gel electrophoresis, will be used to identify the proteins associate with cubilin. By comparing the different pattern between normal and affected dogs, association aspects of the proteins specific to the disease condition can be idetified. The identity of each candidate proteins can be determined by protein microsequencing and database searches. Similar genetic linkage tests will be performed to each candidate gene by lntragenic markers. An alternative approach will be a whole canine genome scan to identify positional candidate genes. 341 highly polymorphic markers of all types have been placed on the 3rd generation dog genome maps with average resolution of 9.0 cM and 95% coverage of the genome. Recently, a high resolution radiation hybrid map with 400 markers has been produced and partially integrated with the linkage map. Informative type I markers will be tested for linkage to canine I-GS. Candidate genes will be identified by comparing the linked region to respective human and mouse genome maps and can be further screened by examining the function and tissue expression of each gene. Candidates will be tested for linkage to the canine disease locus with lntragenic markers. The linked gene identified by either method mentioned above will be cloned and examined for the disease-causing mutations by comparing the gene sequence between the normal and affected dogs. The defined mutation(s) will be further confirmed by checking for the mutation(s) in the normal dog population and functional studies on the gene from both normal and affected dogs. 131 iiiijjjjijjjjjljiiiiii