"Michsgm’a Mate University ——"‘ __— This is to certify that the dissertation entitled COMPARATIVE VIRULENCE OF PORCINE ROTAVIRUS SEROTYPES 1 AND 2 IN GNOTOBIOTIC PIGS presented by JAMES EDWARD COLLINS has been accepted towards fulfillment of the requirements for Ph'D° degreein Pathology Major professor Date July 2, 1986 MSU is an Affirmative Action/Equal Opportunity Institution 0—12771 MSU LIBRARIES V RETURNING MATERIALS: Piace in book drop to remove this checkout from your record. FINES W111 be charged if book is returned after the date stamped below. COMPARATIVE VIRULENCE OF PORCINE ROTAVIRUS SEROTYPES 1 AND 2 IN GNOTOBIOTIC PIGS BY James Edward Collins A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Pathology 1986 '¢-;_ bf; t ‘b "4/ '7'. Th serotyp Dakota determi virus 5 lesion seroty‘ F inocul homoge the Oi Dakot; Totan Morta monit inoeu and 1 the f 928. 8134 ABSTRACT COMPARATIVE VIRULENCE OF PORCINE ROTAVIRUS SEROTYPES 1 AND 2 IN GNOTOBIOTIC PIGS BY JAMES EDWARD COLLINS The purpose of the present study was to compare serotypes 1 (Ohio State University strain) and 2 (South Dakota State University strain) of porcine rotavirus to determine if specific clinical signs, mortality rates, virus shedding patterns, virus antigen distribution, or lesion distribution and severity were related to the serotype of rotavirus with which pigs were infected. Forty hysterotomy derived gnotobiotic pigs were inoculated orally at 3 days of age with 2 ml of homogenate containing 105 pig-infectious dose50 of either the Ohio State University (OSU) strain or the South Dakota State University (SDSU) strain of porcine rotavirus. Controls were inoculated with media only. Mortality, clinical signs, and body weights were monitored daily. Five pigs in each of the virus inoculated groups and 4 control pigs were killed at 24 and 168 hours after inoculation. Four pigs in each of the 3 experimental groups (OSU, SDSU, and control) were killed collect immunof examin; A strain throug SDSU 5 hours obserV OSU st did h< strah but d given immun given after duode and j and j flexi COIIIp.‘ Althl the killed 72 hours after inoculation. Specimens were collected at necropsy for serum chemistry, histologic, immunofluorescent, and scanning electron microscopic examinations. All of the pigs inoculated with the OSU or SDSU strains survived. Control pigs remained healthy throughout the study. Pigs inoculated with the OSU or SDSU strains developed diarrhea 19-48 hours and 24-54 hours after inoculation, respectively. Vomiting was observed in 5 of 14 (36%) of the pigs infected with the OSU strain whereas pigs inoculated with the SDSU strain did not vomit. Pigs inoculated with the OSU or SDSU strains had reduced weight gain compared to control pigs, but there was no difference in weight gain between pigs given the OSU or SDSU strains. Results of immunofluorescent examinations were similar for pigs given either rotavirus strain except that at 24 hours after inoculation, viral antigen was detected at the duodenal-jejunal flexure in 5/5 pigs given the OSU strain and in 1/5 pigs given the SDSU strain. Villous atrophy and fusion were more severe at the duodenal—jejunal flexure of pigs inoculated with the SDSU strain when compared to pigs inoculated with the OSU strain. Although the OSU and SDSU strains did show differences in the occurrence of vomiting and distribution of Villous atrophy, these strains were equally virulent for 3-day—old gnotobiotic pigs. Dedicated, with love, to my wife, Barbara, and our sons, Brian and Daniel. iv Depar Adalb to pu fashi Dr. 8 Dr. E SUPPC stud} Thong C0mmi assi Rita DEmb Dak ACKNOWLEDGEMENTS I wish to express my gratitude to the faculty in the Department of Pathology, particularly Drs. Robert Leader, Adalbert Koestner, and Janver Krehbiel, for allowing me to pursue a doctorate degree in a rather non-traditional fashion. I wish to express my sincere appreciation to Dr. Stuart Sleight, my major professor, and to Dr. David Benfield, my research advisor, for their support, guidance, and friendship during my course of study. I also wish to thank Drs. Roger Maes, Thomas Mullaney, and Glenn Waxler, members of my guidance committee, for their helpful suggestions. Special thanks to Dr. Mahlon Vorhies, Chairman, Department of Veterinary Science, South Dakota State University, for providing financial and personal support for this and several other research projects. I wish to acknowledge Julie Duimstra for technical assistance with electron microscopic studies and Rita Miller for typing this dissertation. I also wish to thank the many faculty and staff members in the Department of Veterinary Science at South Dakota State University who contributed to this project. sons, under: My deepest appreciation to my wife, Barbara, and our sons, Brian and Daniel, for their support, love, and understanding throughout my studies. vi LIST I LIST INTRO LITER MATE TABLE OF CONTENTS Page LIST OF TABLES. . . . . . . . . . . . . . . . . . . . ix LIST OF FIGURES . . . . . . . . . . . . . . . . . . . xi INTRODUCTION. 1 LITERATURE REVIEW . 4 Classification . . . . . . . 4 Rotavirus Serotypes and Subgroups . . . . . 4 . . . . . g 9 Rotavirus Electropherotypes Classification Systems. . Atypical Rotaviruses . . . . . Physicochemical Properties . . . . . . . . . . . 12 Cultivation in Cell Culture. . . . . . . . . . . l3 Morphogenesis. . . . . . . . . . . . . . . l7 Epidemiology . 20 Clinical Signs . . . . . . . . . . . . . . . . . 23 Clinical Pathology . . . . . . . . . . . . . . . 28 Pathophysiology. . . . . . . . . . . . . 30 Lesions and Pathogenesis . . . . . . . . . . . . 31 Gross Lesions . . . . . . . . . . . . 31 Histopathologic Lesions . . . . . . . . . . 32 Scanning Electron Microscopy. . . . . . . . 36 Transmission Electron Microscopy. . . . . . 37 Immunofluorescence and Location of Viral Antigens . . . . . . . . . . . . . 37 Viral Shedding. . . . . . . . . . . . . . . 39 Immunology . . . . . . . . . . . . . . 39 Humoral Immunity. . . . . . . . . . . . . . 39 Cell- mediated Immunity. . . . . . . . . . . 41 Immunization. . . . . . . . . . . . . . . 43 Diagnosis. . . . . . 46 Enzyme— linked Immunosorbent Assays. . . . . 46 Electron Microscopy . . . . . . . . . . . . 48 Immunofluorescence. . . . . . . . . . . . 49 Other Diagnostic Techniques . . . . . . . . 50 Treatment and Control. . . . . . . . . . . . . . 50 MATERIALS AND METHODS . . . . . . . . . . . . . . . . 53 Gnotobiotic Pigs . . . . . . . . . . . . . . . . 53 Virus. . . . . . . . . . . . . . . . . . . 54 RESUl DISC SUI-ll? BIBI VITI‘ Virus Infectivity Titrations . . . . . Experimental Design. . - Clinical Signs and Body Weight . Clinical Pathology . . . Necropsy . Preparation of Fluorescein- conjugated Gamma Globulin. . Examination by Immunofluorescence. Light Microscopy . Scanning Electron Microscopy Electron Microscopic Examination of Feces . . . . Statistical Analysis RESULTS . Clinical Signs Body Weight. . Clinical Pathology . Gross Lesions. Light and Scanning Electron Microscopic Changes . . . . . . . . . . . Immunofluorescence . Electron Microscopic Examination of Feces. DISCUSSION. SUMMARY . BIBLIOGRAPHY. VITA. viii Table LIST OF TABLES Table Page 1 Mean body weights (g 1 SD) for gnotobiotic pigs infected with the OSU or SDSU strains of porcine rotavirus . . . . . . . 69 2 Serum chemistry and hematocrit values for gnotobiotic pigs 24 hours after inoculation with the OSU or SDSU strains of porcine rotavirus. . . . . . . . 71 3 Serum chemistry and hematocrit values for gnotobiotic pigs 72 hours after inoculation with the OSU or SDSU strains of porcine rotavirus. . . . . . 72 4 Serum chemistry and hematocrit values for gnotobiotic pigs 168 hours after inoculation with the OSU or SDSU strains of porcine rotavirus. . . . . . . . 73 5 Villous length (pm) in the small intestines of gnotobiotic pigs infected with the OSU or SDSU strains of porcine rotavirus . . . . 75 6 Crypt depths (um) in the small intestines of gnotobiotic pigs infected with the OSU or SDSU strains of porcine rotavirus. . . . . . 85 7 Light microscopic findings in the small intestine of gnotobiotic pigs 24 hours after inoculation with the OSU or SDSU strain of porcine rotavirus. . . . . . . . 86 8 Light microscopic findings in the small intestine of gnotobiotic pigs 72 hours after inoculation with the OSU or SDSU strain of porcine rotavirus. . . . . . . . . . . . . . 87 9 Light microscopic findings in the small intestine of gnotobiotic pigs 168 hours after inoculation with the OSU or SDSU strain of porcine rotavirus. . . . . . . . . . . . . . 88 ix Table 10 ll 12 Table 10 11 12 Page Immunofluorescence in the small intestine of gnotobiotic piglets infected with the SDSU strain of porcine rotavirus . . . . . . 99 Immunofluorescence in the small intestine of gnotobiotic piglets infected with the OSU strain of porcine rotavirus. . . . . . . . 100 Results of direct electron microscopic examination of feces or cecal content from pigs infected with the SDSU or OSU strain of porcine rotavirus . . . . . . . . . . 102 it... Figur \l Figures 1 10 LIST OF FIGURES Scanning electron micrograph of the UJ of a pig 24 hours after inoculation with the OSU strain of porcine rotavirus Section of small intestine from the MI of a pig 72 hours after inoculation with the OSU strain of porcine rotavirus Section of small intestine from the DU of a pig 72 hours after inoculation with the SDSU strain of porcine rotavirus. Section of small intestine from the DU of a pig 168 hours after inoculation with the SDSU strain of porcine rotavirus. Scanning electron micrograph of the DU of a pig 168 hours after inoculation with the SDSU strain of porcine rotavirus . Section of small intestine from the DU of a pig 168 hours after inoculation with the OSU strain of porcine rotavirus. Scanning electron micrograph of the DU of a pig 168 hours after inoculation with the OSU strain of porcine rotavirus. Villous fusion in a section of small intestine from the LJ of a pig 72 hours after inoculation with the SDSU strain of porcine rotavirus . . . . . . Scanning electron micrograph of small intestine from the LJ of a pig 168 hours after inoculation with the SDSU strain of porcine rotavirus Villous fusion in a section of UJ from a pig 168 hours after inoculation with the OSU strain of porcine rotavirus. xi Page 77 78 79 80 81 82 83 89 9O 91 Figur ll 12 l3 14 IS Figures 11 12 l3 14 15 Most of the absorptive cells lining villi in the LI of a control ' 24 hours after inoculation with media are vacuolated (+4) . . . . Immature cuboidal to low columnar absorptive cells line an atrophic villus in the UJ of a pig 24 hours after inoculation with the OSU strain of porcine rotavirus. Duodenum of a control pig killed at post- -inocu1ation hour 24. . Scanning electron micrograph of the duodenum of a control pig at post—inoculation hour 24. . Scanning electron micrograph of the lower ileum of a control pig at post- inoculation hour 24 . . . . Page 92 94 95 96 97 ,3 healt sheer Stri< Mebu: in c; viru. impO' mamm eta chil etio thro for al Were the You whi. dis deS INTRODUCTION Rotaviruses were first identified in the feces of a healthy vervet monkey and in intestinal washings from sheep and cattle at a slaughterhouse (Malherbe and Strickland-Cholmley, 1967). However, it was not until Mebus et a1 (1969) associated a rotavirus with diarrhea in calves in Nebraska that the significance of these viruses was recognized. Rotaviruses are now considered important causes of neonatal gastroenteritis in many mammalian and avian species (Mebus et al, 1971b; Bishop et al, 1973; Bohl et a1, 1978; McNulty et al, 1978). In children, rotavirus is the single most important etiologic agent of acute infantile gastroenteritis throughout the world and has been estimated to account for one million diarrheal deaths annually (Vesikari et al, 1983). Initially, all viruses with rotavirus morphology were thought to possess a common group antigen located on the inner capsid layer of the virus (Woode et al, 1976a; Yolken et al, 1978a). Recently, several rotaviruses which are morphologically similar to but antigenically distinct from the conventional rotaviruses have been described. These viruses have been referred to as parar virus (Snod isola typic the p group et a] base< Serow caps: dete- neut' 1984 are 1976 immu fixa 1980 subg SQVE and I‘ICN1 muCl pararotaviruses (Bohl et a1, 1982), rotavirus-like viruses (Rodger et a1, 1982), and atypical rotaviruses (Snodgrass et a1, 1984). To distinguish among these isolates, Pedley et a1 (1983) proposed a scheme in which typical and atypical rotaviruses are classified based on the presence of distinctive group specific antigens into groups designated A, B, C, and recently D and E (Pedley et al, 1986). Group A rotaviruses have been further classified based on serotype and subgroup specific antigens. Serotype specific antigens are located on the outer capsid layer of the virus (Bridger, 1978), and are detected by plaque reduction and fluorescent focus neutralization assays (Sato et a1, 1982; Bohl et a1, 1984; Hoshino et a1, 1984). Subgroup specific antigens are located on the inner capsid layer (Woode et al, 1976a) and are detected by enzyme—linked immunosorbent, immune-adherence hemagglutination, and complement fixation assays (Struker et a1, 1979; Zissis and Lambert, 1980). By using these assays, multiple serotypes and subgroups of mammalian rotaviruses have been identified. Recent reports have indicated differences in clinical signs, viral antigen distribution, and lesion severity for different rotavirus serotypes, subgroups, and groups. An avian group A rotavirus, studied by McNulty et a1 (1983) had a predilection for the duodenal mucosa whereas a non—group A avian rotavirus replicated ider cro: 916 al, pre Sig '_.J 98 $91“ We 1; best in the mid-small intestine. In gnotobiotic pigs, a group B porcine rotavirus caused mild clinical signs and lesions (Theil et a1, 1985) whereas a group A porcine rotavirus (Ohio State University strain) caused severe diarrhea and extensive intestinal damage (Theil et a1, 1980). Four strains of rotavirus isolated from diarrheic calves caused different amounts of Villous atrophy when inoculated into ligated intestinal loops in colostrum deprived calves (Carpio et al, 1981b). Differences in the clinical symptoms associated with infection by two subgroups of rotavirus in humans have been reported by some investigators (Uhnoo and Svensson, 1986), but these findings are in disagreement with the findings of others (White et a1, 1984). Two serotypes of porcine rotavirus have been identified by plaque reduction neutralization tests, cross protection studies in gnotobiotic piglets, and electrophoresis of rotaviral double-stranded RNA (Bohl et a1, 1984; Hoshino et a1, 1984). The purpose of the present study was to determine if specific clinical signs, mortality rates, virus shedding patterns, and lesion distribution and severity were related to the serotype of porcine rotavirus with which gnotobiotic pigs were infected. simi bu03 doul Rot.- reo‘ gem See] Sim Wit reo n she LITERATURE REVIEW Classification Rotaviruses derive their name from the Latin word "rota" meaning wheel, which they resemble in appearance (Flewett et a1, 1974). Initially, they were referred to as "orbivirus", ”duovirus”, "reovirus-like agent" and "infantile gastroenteritis virus" (Mebus et al, 1971b; Bishop et a1, 1973; Kapikian et a1, 1975). Rotaviruses, reoviruses, and orbiviruses have many similarities. They are approximately the same size (65 to 75 nm), have double-shelled capsids, have similar buoyant density in cesium chloride and possess a double-stranded RNA genome (Palmer et a1, 1977). Rotaviruses can be distinguished from orbiviruses and reoviruses by the presence of 11 segments of the RNA genome; orbiviruses and reoviruses possess only 10 segments (Palmer et a1, 1977). On the basis of these similarities and differences, rotaviruses are classified with reoviruses and orbiviruses within the family reoviridae (Matthews, 1979). Rotavirus Serotvpes and Subgroups Most rotaviruses from mammalian and avian species share a common antigen that is associated with the inner capsi antig locat rotat adapt and 1 devei rota1 seve: and . I983 capsid layer (Woode et al, 1976a). Serotype specific antigens are distinct from the common antigen and are located on the outer capsid (Bridger, 1978). When rotaviruses from several mammalian species became adapted to replicate in cell culture, plaque reduction and fluorescent focus neutralization assays were developed for the antigenic comparison of different rotaviruses. These assays led to the recognition of several serotypes of rotaviruses from different mammalian and avian species (Sato et a1, 1982; Murakami et a1, 1983; Bohl et a1, 1984; Hoshino et a1, 1984). In addition to neutralization assays, rotaviruses have been studied by enzyme-linked immunoassays, complement fixation, immune—adherence hemagglutination, and other techniques (Struker et a1, 1979; Zissis and Lambert, 1980). The use of these assays led to much confusion in the literature about the serologic classification of rotaviruses because it was assumed that the above techniques gave identical results to conventional neutralization assays. It has been shown, however, that antigens detected by enzyme immunoassays, immune—adherence hemagglutination, and complement fixation are distinct from those detected by plaque reduction or fluorescent focus neutralization (Kapikian et a1, 1981). ”Serotypes" as determined by neutralization tests depend on differences in polypeptides located on the outer capsi enzyme-1mm hmmne-adh difference et al, 19% proposed designati "subgroup differenc fixation et al, 1 A c and dive rotaviri 1984). from se Studied antiger basis t neutra antlse IOtavj human serot- an eq serOt hoshj outer capsid whereas ”serotypes" as determined by enzyme-immunoassays, complement fixation, and immune-adherence hemagglutination are based on differences in the major inner capsid polypeptide (Kalica et a1, 1981). To avoid confusion, Kapikian et a1 (1981) proposed that the term "serotype" be reserved for designation of neutralization specificity, and the term "subgroup" be used in place of serotype, to indicate differences detected by enzyme immunoassays, complement fixation, and immune-adherence hemagglutination (Kapikian et a1, 1981). A comprehensive study of the serotypic similarity and diversity of human and other mammalian and avian rotaviruses has been reported recently (Hoshino et a1, 1984). Sixteen different strains of rotavirus derived from seven mammalian species and two avian species were studied by plaque reduction neutralization. Seven antigenically distinct serotypes were established on the basis of a greater than 20-fold difference between neutralizing titers of homologous and heterologous antiserum. In this study, three strains of porcine rotavirus (Gottfried, SB-lA and SB—Z), and one strain of human rotavirus (St. Thomas No. 4) were of the same serotype designated type 4. Porcine rotavirus (OSU) and an equine rotavirus (H—l) made up a possible fifth serotype. The serotype 4 rotaviruses as described by Hoshino et a1 (1984) represent an example of shared serotype and thos The OSU stre subgroup Strain E I, wher: sungOU' example are of specifi Bc rotavir with dj identi: rossa electr. Ohio 8 as the Strain SerOty rotavi t0 se1 HoshiI Et al in thl serotype specificity between rotaviruses of human origin and those from other animals. The Gottfried strain of porcine rotavirus and the OSU strain of porcine rotavirus were found to have subgroup II and subgroup I specificities, respectively. Strain SB-Z of porcine rotavirus was found to be subgroup I, whereas the Gottfried strain, as mentioned above, was subgroup II. This pair of isolates represents the first example of two rotaviruses from one animal species that are of the same serotype, but differ in subgroup specificity. Bohl et a1 (1984) studied seven strains of rotaviruses isolated from intestinal contents of piglets with diarrhea. Two serotypes of porcine rotavirus were identified by plaque reduction neutralization tests, cross—protection studies on gnotobiotic piglets, and electrophoresis of rotaviral double-stranded RNA. The Ohio State University (OSU) porcine isolate was suggested as the prototype serotype 1 rotavirus. The Gottfried (G) strain porcine rotavirus was suggested as the prototype serotype 2 rotavirus. Serotypes l and 2 porcine rotaviruses as described by Bohl et a1 (1984) correspond to serotypes 5 and 4, respectively, as described by Hoshino et a1 (1984). The classification system of Bohl et a1 (1984) for porcine rotavirus strains will be used in the remainder of this dissertation. It systems based c polyacr (1981) electrt segmen are dm 7-9 as Differ within la, 11 classi conduc I rotavi subgrc rOtavi t0 sul 10 an< Subgn apply Elecp Of my disti 1982, Rotavirus Electropherotypes In addition to subgroup and serotype classification systems, there has been an effort to classify rotaviruses based on the mobility of their RNA segments during polyacrylamide gel electrophoresis. Lourenco et a1 (1981) have proposed a scheme for rotaviral electropherotype classification in which the 11 RNA segments are divided into four groups. RNA segments 1—4 are denoted as group I, bands 5 and 6 as group II, bands 7-9 as group III, and bands 10 and 11 as group IV. Differences in the relative migration of RNA segments within a group are designated by a small letter (example Ia, IIa, IIIb, IVc). This system allows rapid classification of RNA patterns when standard methods of conducting the test are employed. The migration of RNA segments 10 and 11 of human rotavirus strains has been shown to correlate with subgroup specificity (Kalica et al, 1981). Human rotaviruses with slow migrating segments 10 and 11 belong to subgroup I, whereas those with fast migrating segments 10 and 11 belong to subgroup II. Such a correlation of subgroup specificity with RNA electropherotype does not apply to animal rotaviruses. Comparison of RNA electropherotypes has proved useful in the identification of new groups of rotaviruses that are antigenically distinct from the classical rotaviruses (Bohl et a1, 1982; Bridger et a1, 1982; Snodgrass et a1, 1984). Classii A rotavi: scheme Graham serolo crypto distin source need u that r at leg or 9 a from Commc Capsi a1 micrt indl.‘ Classification Systems A unified serological classification for rotaviruses, similar to the universal classification scheme for influenza virus, has not yet been achieved. Graham and Estes (1985) recently have proposed a serologic classification system which utilizes a cryptogram to convey the information necessary to distinguish and identify new rotavirus isolates from any source. However, this classification system may already need modification. Greenberg et a1 (1983) demonstrated that neutralization of a simian rotavirus is mediated by at least two gene products of rotavirus; those of gene 8 or 9 and gene 4. Rotavirus genes 8 or 9 code for the major neutralization antigen (VP7) whereas gene 4 codes for a minor neutralization antigen (VP3). Hoshino et al (1984) suggested that future systems of rotaviral classification may need to indicate two, instead of one, distinct serotypic specificities for each isolate of rotavirus. Atypical Rotaviruses Initially, all viruses with rotavirus morphology from different animal species were thought to possess a common group antigen or antigens located in the inner capsid layer of the virus (Woode et al, 1976a; Yolken et a1, 1978). In 1980, Saif et a1 detected, by electron microscopy, virus particles that were morphologically indistinguishable from rotaviruses but which did not cross-1 rotavii infecti particl seroty] finding type. this ”i (Bohl R which antige referr rotavi rotavi A became antlge t0 eac a Clas Virus; SChEme Class: subcli SPEci; Additj SpeCi: 10 cross—react with antisera to porcine, bovine, or human rotaviruses or to reovirus type 3. Furthermore, previous infection of gnotobiotic pigs with the rotavirus-like particles failed to protect them from challenge with serotype 1 porcine rotavirus (strain OSU). These findings suggested the presence of a ”new" rotavirus type. The pathogenesis of rotaviral diarrhea caused by this "new rotavirus” was detailed in a subsequent report (Bohl et a1, 1982). Recently, several additional atypical rotaviruses which lack the conventional rotavirus group specific antigen have been described. These viruses have been referred to as pararotaviruses (Bohl et a1, 1982), rotavirus-like viruses (Rodger et a1, 1982), and atypical rotaviruses (Snodgrass et a1, 1984). As these viruses were studied in greater detail, it became evident that several atypical rotaviruses were antigenically distinct from and biochemically unrelated to each other. Therefore, it became necessary to develop a classification scheme to distinguish among these viruses. Pedley et a1 (1983) proposed a classification scheme in which typical and atypical rotaviruses were classified in the genus as rotaviruses but further subclassified, based on the presence of distinctive group specific antigens into groups designated A, B, and C. Additional atypical rotaviruses that do not possess group specific antigen characteristic of groups A, B, or C have been 1 groups Pedley l rotav: for g enzym nongr antis is re rotav also (Pedl elect group Where doubi in d unkn rota al idem f8ce Davi in t 11 been identified indicating the need for additional groups (Snodgrass et a1, 1984; Vonderfecht et a1, 1984; Pedley et a1, 1986). Detection of samples which are positive for rotavirus particles by electron microscopy but negative for group A rotavirus in fluorescent antibody or enzyme—linked immunoassay tests is suggestive of a nongroup A rotavirus. But further testing, using antisera which reacts with only the subgroup in question, is required to confirm the presence of atypical rotaviruses (Pedley et a1, 1983). Electrophoresis of viral RNA in polyacrylamide gel also is helpful in distinguishing rotavirus groups (Pedley et a1, 1983). The major difference in the RNA electrophoretic profiles of groups A, B, and C is that group A rotavirus bands 7, 8, and 9 migrate as a triplet, whereas in groups B and C, this triplet is replaced by a doublet. At present, the importance of atypical rotaviruses in diarrheal diseases of mammalian and avian species is unknown. It appears that the prevalence of atypical rotaviruses in cattle and swine is quite low (Chasey et a1, 1984; Theil et a1, 1985). Atypical rotavirus are identified more frequently than group A rotaviruses in feces from diarrheic turkey poults and lambs (Chasey and Davies, 1984; Saif et a1, 1985) so they are of importance in those species. I of sex and Sc agree: does 1 gener. types Negat viewe smoot ”fuzz almos IOtav diame C8811 aPPT< Chlo: al, j been Rota solv Physicochemical Properties The structure of the rotavirion has been the subject of several investigations (Martin et a1, 1975; Stammond and Schoub, 1977). Although there is no general agreement about the number of capsomeres, the rotavirion does have a definite capsomere structure. It is generally agreed that rotaviruses have two main particle types: single— and double—shelled (see Morphogenesis). Negatively stained double—shelled virus particles, as viewed with the transmission electron microscope, have a smooth outer layer. In contrast, the orbiviruses have a "fuzzy" outer layer, and the outer capsid of reovirus is almost featureless (Palmer et a1, 1977). Double-shelled rotavirus particles are approximately 70 to 75 nm in diameter and have a buoyant density of 1.36 g/ml in cesium chloride. Single—shelled particles are approximately 65 nm in diameter with a density in cesium chloride of 1.38 g/ml (Bridger and Woode, 1976; Rodger et a1, 1975). The physicochemical properties of rotaviruses have been discussed in detail (Flewett and Wood, 1978a). Rotaviruses are stable to non—ionic detergents, lipid solvents, heat, extremes of pH and high salt concentrations. Such stability is central to the survival of rotaviruses in the intestinal tract. Simian rotavirus SA—ll is rapidly inactivated when heated at 50°C in the presence of 2 M MgClZ, but is stabilized by heatin also s 1981). rotavi from t rendei l disin and c (752) reduc organ wides (I977 conte disit rota; Cond: habi adap SA—l asyu res; rOte Stic 13 heating in 2 M MgSO4 (Estes et a1, 1979). Calcium ions also stabilize rotavirus infectivity (Shirley et a1, 1981). Chelators such as EDTA may be used to destabilize rotavirions by removing divalent cations such as calcium from the outer coat of the virus particle, thereby rendering them more permeable (Cohen, 1977). Rotaviruses are resistant to many chemical disinfectants and antiseptics commonly used in research and chemical laboratories (Sattar et a1, 1983). Ethanol (752) and formaldehyde (3.7%) are somewhat effective in reducing the amount of virus even in the presence of organic matter, but these chemicals are impractical for widespread use in a farm environment. Snodgrass et a1 (1977) found a rotavirus purified from the intestinal contents of a lamb resistant to iodine-based disinfectants. Because of the resistant nature of rotaviruses, it is unlikely that disinfection under farm conditions will be completely successful. Cultivation in Cell Culture Rotaviruses grow extremely well in their natural habitat, the intestinal tract, but have been difficult to adapt to cell culture. The first rotaviruses cultured, SA—ll and the ”0" agent, were isolated from an asymptomatic vervet monkey and slaughterhouse waste, respectively. These viruses were not recognized as rotaviruses until much later (Malherbe and Stickland—Cholmley, 1967). Interest in the cultivation of rot virus SUCCEE 19713} the isol hung Cont Feh‘ to} 14 of rotaviruses intensified when neonatal calf diarrhea virus (NCDV), a cause of bovine calf diarrhea, was successfully isolated in cell culture (Mebus et al, 1971a). Excluding bovine rotavirus, serial propagation of these viruses in cell culture proved difficult (Woode et al, 1976b; McNulty et al, 1976a). Porcine rotavirus was successfully passaged in cell culture by treatment of viral suspensions with pancreatin prior to inoculation onto cell monolayers (Theil et al, 1977). In a subsequent paper, Theil et a1 (1980) reported that rotavirus infectivity for porcine kidney cells (PK—15) was enhanced by incorporation of pancreatic endopeptidases into the cell culture maintenance medium. These important findings were widely applied and led to the successful cultivation of rotaviruses derived from many mammalian and avian species (McNulty et a1, 1979; Wyatt et a1, 1980; Tajima et a1, 1984; Makabe et a1, 1985). It has not proved possible, however, to culture all rotaviruses by these methods. Rotaviruses vary in their requirement for trypsin in the culture medium. The SA-ll and bovine rotavirus isolates (NCDV) were cultured without trypsin whereas human rotavirus has only been isolated with culture media containing trypsin (Wyatt et al, 1980; Sato et a1, 1981). Feline, canine, and simian rotavirus strains were found to be less dependent upon trypsin than human, bovine, porcin 1981). not re strait cell < virus 15 porcine, chicken, and turkey rotaviruses (Hoshino et a1, 1981). Although trypsin treatment of bovine rotavirus is not required for the isolation of some bovine rotavirus strains, it can effectively enhance viral replication in cell culture, thus producing high titer stocks of the virus (Clark et a1, 1979). The molecular basis for the proteolytic enhancement of rotavirus infectivity has been studied (Espejo et a1, 1981; Estes et al 1981). Trypsin cleaves a major non—glycosylated polypeptide of the outer capsid (VP3) yielding two polypeptides with molecular weights of approximately 60,000 and 28,000. It is proposed that the latter polypeptides contain hydrophobic regions that, once exposed, aid in viral penetration into the host cell. Primary kidney cell cultures from a variety of species have been the cell—type most frequently used to culture rotaviruses. At present, a number of cell lines are being used successfully, including MDBK (Madin-Darby bovine kidney), PK-lS (porcine kidney), BSC—l (green monkey kidney), LLC—MK2 (Rhesus monkey kidney), CV-l (green monkey kidney), and MA—104 (Rhesus monkey kidney) (Wyatt and James, 1982). Primary cells appear to support virus growth more efficiently than continuous cell lines and therefore are more sensitive for rotavirus isolation (Ward et a1, 1984). Rotavirus-specific antibodies are prevalent in the sera c 1981), use of Rotavj serum inter: or an‘ part, in se cultu tryps diarr inocr (Welt deter hours VaCUt incl cres smal The of C bovi iDOC l6 sera of domestic and laboratory animals (Sato et a1, 1981), so for cultivation investigators must avoid the use of adult animal sera containing rotavirus antibody. Rotavirus antibodies have been detected in fetal bovine serum and in purified serum albumin preparations and may interfere with diagnostic assays for rotavirus antigens or antibodies (Offit et a1, 1984). This finding, in part, explains why human rotavirus has been isolated only in serum-free media. Cytopathic effect (CPE) caused by rotavirus in cell culture depends on the cell-type used, the presence of trypsin, and the strain of rotavirus. Neonatal calf diarrhea virus produces an easily discernible CPE when inoculated at high concentrations onto cell cultures (Welch and Twiehaus, 1973). Cytopathic effect is detectable by 24 hours after inoculation, and by 48 hours, some areas of the cell culture may be devoid of cells. Infected cells may appear granular and finely vacuolated, and contain single or multiple eosinophilic inclusions. Some cells become thin, elongated, and crescent-shaped and remain attached to the monolayer only by a single process. Those that completely detach are small, rounded or elongated and have pyknotic nuclei. The cytopathic effect has been produced in a wide variety of cell culture systems (Fernelius et a1, 1972). Another bovine rotavirus strain produces similar CPE when inoculated onto MDBK kidney cells, except a larger portior inclusi II rotavi1 culture produc: the de‘ Plaque medium other inocul a plaq for as widely rotavi I984), l7 portion of the monolayer is destroyed, and cytoplasmic inclusions are uncommon (McNulty et a1, 1977). In contrast to neonatal calf diarrhea virus, porcine rotavirus strains produce minimal to moderate CPE in cell culture unless the strain has been passaged several times 13 yipgg. The detection of rotavirus strains that produce minimal CPE in cell culture has been improved by the development of plaque assays (Bohl et a1, 1984). Plaque assays utilize pancreatin or trypsin in an overlay medium. In this system, porcine rotavirus, as well as other rotavirus strains, will produce plaques after inoculation onto MA-104 cells. By use of this technique, a plaque reduction neutralization test has been developed for assaying neutralizing antibody and has been used widely for the detection of antigenic diversity among rotavirus strains (Bohl et a1, 1984; Hoshino et a1, 1984). Morphogenesis The morphogenesis of rotavirus has been studied by electron microscopic examination of cell cultures (Chasey, 1977; Saif et a1, 1978; Quan and Doane, 1983; Suzuki et a1, 1984) and intestinal epithelial cells (Chasey, 1977; Pearson and McNulty, 1979). The morphogenetic stages of rotaviruses isolated from different animal species is similar both in giggg and 13 yiyg. Because of these similarities as well as the early unsuccessful attempts to isolate human rotavirus strains, tissue-cultua model system (Quan and Do Study 0 culture reve virions ente (b) in some into lysosou found first as 6 hours a cytoplasmic viroplasm b' these part1 enveIOpe, w thus formin lysis. DeSpit morPhogene: the replic. (1985) hay 104 cells PTEtreatme CYtoplasm dissolutic abSence of Virions we V. lrug att.‘ 18 tissue-culture-adapted simian rotavirus SA-ll became a model system for the study of rotavirus morphogenesis (Quan and Doane, 1983). Study of the morphogenesis of SA-ll virus in cell culture revealed the following sequence of events: (a) virions enter the cell by receptor—mediated endocytosis; (b) in some instances, the virus is taken up directly into lysosomes, but in others, the virus particles are found first in endosomes and then lysosomes; (c) as early as 6 hours after infection, immature virus can be seen in cytoplasmic aggregates of viroplasm and at the edge of viroplasm budding into rough endoplasmic reticulum; (d) these particles become enveloped during budding, but the envelope, which is not required for infectivity, is lost, thus forming a mature virion which is released by cell lysis. Despite the many reports about rotavirus morphogenesis, some aspects concerning early stages of the replication cycle are controversial. Suzuki et a1 (1985) have found that the mode of viral entry into MA 104 cells is affected by trypsin. With trypsin pretreatment, viral nucleoids passed directly into the cytoplasm within 5 minutes after inoculation through dissolution of viral capsid and cell membrane. In the absence of trypsin, phagocytosis occurred in which virions were sequestered into lysosomes 20 minutes after virus attachment to cell membranes. After sequestration, uncoating of but it did n Therefore, S phagocytosis The cap least 5 proi 1W2, VP6) m; (VP3 and VP Estes et a1 identified inclusions believed ti assembled f Outer sheli Process (P. multivesic morPhogene (SuzUki 8t Multi identifiec Petrie et SiHSIE-sh. cells, bu Particles NOQde, 19 CYtOplaSK a1. 1981, l9 uncoating of rotavirus virions within lysosomes was seen, but it did not result in the release of the viral genome. Therefore, Suzuki et a1 (1985) discounted the theory that phagocytosis is related to viral replication. The capsid of simian rotavirus SA-ll consists of at least 5 protein classes. Three of these proteins (VPl, VP2, VP6) make up the inner layer, whereas the other two (VP3 and VP7) form the outer layer (Espejo et a1, 1981; Estes et a1, 1981). Immunocytochemistry studies have identified VP2 and VP6 associated with viroplasmic inclusions (Petrie et a1, 1982). Therefore, it is believed that the core and inner capsid layer are assembled in the viroplasmic inclusions and that the outer shell glycoprotein is added during the budding process (Petrie et a1, 1982). Tubules, fibrils, and multivesicular bodies have been associated with rotavirus morphogenesis, but their origin and function are unknown (Suzuki et a1, 1984). Multiple rotaviral particle types have been identified (Saif et a1, 1978; Pearson and McNulty, 1979, Petrie et al, 1981). Double—shelled particles and single—shelled particles are released from infected cells, but only the smooth double—shelled rotavirus particles have been found to be infectious (Bridger and Woode, 1976). Two additional particle types in cytoplasmic organelles have been described by Petrie et a1, 1981. The first is a subviral particle which is the uncoated vir enveloped pa Enveloped pe released frc in negative? examined wii (1972) did . negatively monkey, but contaminati Serolc antibodies mammalian 2 et al, 197% Bohl et 31 samples an serologica IOtaviruS. also has b 31, 1984; antibody t common in 1958)» bin pIEValenC, Ohio SWin indiCate 20 uncoated virion seen within lysosomes. The second is an enveloped particle seen in the endoplasmic reticulum. Enveloped particles lose their envelopes prior to being released from the endoplasmic reticulum so are not found in negatively stained preparations of fecal material when examined with an electron microscope. E13 and Lecatsas (1972) did observe enveloped virus particles in negatively stained preparations from a healthy vervet monkey, but this is unusual and may have represented contamination by another virus. Epidemiology Serologic studies on the prevalence of rotavirus antibodies indicate a widespread distribution in many mammalian and avian species (Petri et a1, 1978; McNulty et a1, 1978; Bohl et a1, 1984; Bridger and Brown, 1985). Bohl et a1 (1984) found that 94% of 274 porcine serum samples and 1002 of 75 herds in the United States were serologically positive to the OSU strain of porcine rotavirus. A high prevalence of antibodies to rotavirus also has been found in swine in other countries (Utera et a1, 1984; Bridger and Brown, 1985). The prevalence of antibody to the atypical rotaviruses (groups B and C) is common in pigs in the United Kingdom.(Bridger and Brown, 1958), but Theil et a1 (1985) found a much lower prevalence (23%) of antibody to group B rotaviruses in Ohio swine sera. Results of serologic surveys also indicate that some serotypes of human rotavirus are more prevalent th least two se distinguishe comparing tl rotavirus t} There a many infect: is more pre‘ et a1, 1976 is not know suggested a the and Shi feces on er at high or humidity, rotavirus : DEER Sugge: be a refle- numbers, TO as transmissi Comparison diffErent infectiOn 21 prevalent than_others (Yolken et a1, 1978). Although at least two serotypes of porcine rotavirus have been distinguished (Bohl et a1, 1984), epidemiologic surveys comparing the prevalence of infection by porcine rotavirus types 1 and 2 have not been reported. There are seasonal variations in the prevalence of many infectious diseases. Rotaviral diarrhea in people is more prevalent in the fall and winter months (Kapikian et a1, 1976). Although the cause of the seasonal pattern is not known, low relative humidity in the homes has been suggested as a factor influencing rotavirus survival. Moe and Shirley (1982) found that rotaviruses in feces on environmental surfaces survive for days or weeks at high or low relative humidity, but not intermediate humidity. A seasonal increase in the prevalence of rotavirus infections in animals during the winter has been suggested (Woode and Bridger, 1975a), but this may be a reflection of seasonal variation in livestock numbers. To assess the importance of interspecies transmission of rotaviruses and to make antigenic comparisons between rotavirus strains isolated from different species, numerous experimental cross—species infection studies have been done (Mebus et a1, 1977; Tzipori et a1, 1980; Bridger and Brown, 1984). Some strains of rotavirus isolated from children with diarrhea readily infected pigs (Middleton et a1, 1975). Woode and Bridger (197 infection by investigate: rotavirus i: Many 0 infection b‘ recognition discrepanci human or ot animals may in these e) have shown infect pig: porcine ro' PP-l) that rotavirus relationsh because, i r0tavirus CTOSS-neut 81.1ng of rotavi] et 81 (19: are SUSCe‘ rotavirus Alth aanSt ev 22 Bridger (1975a) reported that calves were refractory to infection by a strain of human rotavirus, but other investigators successfully infected calves with a rotavirus isolated from people (Mebus et a1, 1977). Many of the investigations of cross-species infection by rotaviruses were completed prior to the recognition of multiple rotavirus serotypes. Thus, discrepancies in the literature concerning the ability of human or other animal rotaviruses to infect experimental animals may be related to the use of different serotypes in these experiments. Bridger and Brown (1984) recently have shown that the ability of bovine rotaviruses to infect pigs depends on the antigenic relationship to porcine rotaviruses. Only a bovine rotavirus (strain PP-l) that was antigenically closely related to porcine rotavirus caused disease. However, a close antigenic relationship does not imply similarities in Virulence because, in the same report, another strain of bovine rotavirus (CP—l) which was indistinguishable from PP-l by cross-neutralization tests, was not pathogenic for pigs. Subgroup characteristics also influence the ability of rotaviruses to infect atypical host species. Zissis et a1 (1983) have shown that colostrum-deprived piglets are susceptible to several human—origin strains of rotavirus, except those belonging to subgroup I. Although rotaviruses are widely distributed in almost every species, there is no evidence that under natural cone Cross-specie has enabled models for ‘ Simult. electropher' detected (8 Bohl et a1, such "mixed known prOpe occur. Th1 variation 5 continued p antigenic \ rotaviruses (Gething 9 among rota‘ reported r The c described gnotobioti 23 natural conditions, animal rotaviruses infect people. Cross-species infection is important, however, because it has enabled the development of many laboratory animal models for the study of rotavirus infections. Simultaneous infection by more than one electropherotype of human or porcine rotavirus has been detected (Spencer et a1, 1983; Rodriguez et a1, 1983; Bohl et a1, 1984). It has been postulated that during such "mixed” infections, genetic reasssortment, a well known property of rotaviruses (Greenberg et a1, 1981) may occur. This genetic reassortment could lead to antigenic variation among rotaviruses, thus enabling their continued persistence within a population. Constant antigenic variation by influenza viruses which, like rotaviruses, have a segmented genome, is well recognized I! (Gething et a1 1980), and evidence of antigenic drift" among rotavirus isolates from human neonates has been reported recently (Coulson et a1, 1985). Clinical Signs The clinical signs of rotavirus infection have been described in conventional, colostrum~deprived, and gnotobiotic animals of many species (Theil et a1, 1978; McAdaragh et a1, 1980; Johnson et a1, 1983; McNulty et a1, 1983; Gillespie et a1, 1984). The first clinical signs, anorexia and reluctance to move, are seen in gnotobiotic piglets within 12—36 hours after inoculation (Crouch and Wood, 1978; Theil et a1, 1978). Similar incdbation 1 species inc: is usually : which time ' Vomiti has been re McAdaragh e 1980). Var used to exp the discrep vomiting 11 been repor- subgroup o isolate ca 1 rotaviru 81, l978b) that clini severe in I rOtavin difference among Chi Ther mucosa is 1977), 24 incubation periods have been reported in several other species including people (Yolken et al, 1978b). Anorexia is usually resolved by 72 hours after inoculation at which time piglets resume eating. Vomiting by piglets infected with porcine rotavirus has been reported inconsistently (Theil et a1, 1978; McAdaragh et a1, 1980; Torres-Medina and Underdahl, 1980). Variations in the serotypes of porcine rotavirus used to experimentally infect the piglets might explain the discrepancies in reports of vomiting. For example, vomiting in people with rotaviral gastroenteritis has been reported to be influenced by the serotype or subgroup of rotavirus involved. A Type 2 human rotavirus isolate caused vomiting in 9 of 16 patients whereas Type 1 rotavirus caused vomiting in only 3 of 11 (Yolken et a1, l978b). Uhnoo and Svensson (1986) recently reported that clinical symptoms (vomiting and diarrhea) were more severe in people infected with subgroup II than subgroup I rotaviruses. However, White et a1 (1984) found no differences in the occurrence of fever or vomiting among children shedding different subgroups of rotavirus. There has been no clear evidence that the gastric mucosa is infected by rotavirus (Pearson and McNulty, 1977). Apparently, injury to the small bowel mucosa is sufficient to initiate a reflex resulting in vomiting. Vomiting is usually not a feature of field cases of porcine rotavirus infection (Bohl et al, 1978) a finding that is some from transm. seen in rot 1977) but h in human in Rotavi acute loss days follow inoculatior expected we Mortai variable. contents 0; exPeriment did not co (Woode et rotavirus reported ( affected b management (1978) for Conditions increased p188. It Well fed, Subc (Cr00ch a. that is sometimes useful in differentiating this disease from transmissible gastroenteritis. Fever is usually not seen in rotaviral infection in pigs (Pearson and McNulty, 1977) but has been associated with rotaviral infections in human infants and young children (Carr et al, 1976). Rotavirus infection in gnotobiotic pigs may cause acute loss of body weight (lo-40%) over the first 2—3 days followed by rapid recovery. At 5—7 days after inoculation, pigs may still be below (20-432) their expected weight (Woode, 1979). Mortality rates caused by porcine rotavirus are variable. When gnotobiotic piglets were inoculated with contents of the same vial of porcine rotavirus in two experiments, mortality rates varied from 0-13Z and this did not correlate with the dose of Virus inoculated (Woode et al, 1976b). In field cases of porcine rotavirus infection, mortality rates of 7-152 were reported (Bohl et al, 1978). Mortality rates are affected by environmental, nutritional, and other management factors. For example, Crouch and Woode (1978) found that, under natural or experimental conditions, a drop of 10 to 20°C in ambient temperature increased the mortality associated with the disease in pigs. It appears that if pigs are kept warm, dry, and well fed, mortality can be reduced. Subclinical infections by rotavirus are common (Crouch and Woode, 1978; Banatvala et al, 1978), and the severity of infections a part, age-de 1002 mortali lOZ mortalit age. The c] also are age age do not < (Sheridan e‘ also occur cattle (Cro some instan gastroenter instances c 1979). Pigs l disease cat infection. exPlain th foufid youn adults bee e16€trolyt not Younge logs from absorptior include t1 intestine 26 severity of clinical signs in experimental rotavirus infections are quite variable. This variation is, in part, age-dependent. Woode et al (1976b) noted up to 100% mortality in piglets infected at 0-4 days of age, 10% mortality at 5-7 days, and no mortality at 28 days of age. The clinical signs of rotavirus infection in mice also are age—dependent. Mice greater than 10—14 days of age do not develop clinical signs of rotavirus infection (Sheridan et al, 1983). Subclinical rotavirus infections also occur in adult swine (Benfield et a1, 1982) and cattle (Crouch and Acres, 1984). Human rotaviruses, in some instances, may affect adults causing severe gastroenteritis (Echeverria et a1, 1983), but in other instances only mild infections occur (Wenmann et al, 1979). Pigs have a remarkable age—resistance to clinical disease caused by transmissible gastroenteritis infection. A number of explanations have been offered to explain this age-resistance. Cornelius et a1 (1968) found young pigs more vulnerable to dehydration than adults because of changes in body water content and renal electrolyte regulation with age. The colon of older, but not younger, pigs can compensate for the increased fluid loss from the small intestine by increased fluid absorption (Argenzio et a1, 1984). Other explanations include the greater involvement of the upper small intestine and the slower proliferation of the intestinal crypts resul aborptive ep One or more age-resistar age-depender difference : enterocytes Riepenhoff—' receptors 0' than in mic does not co susceptibil age are ref infected, 2 Similar to diarrhea (1 mice to re: ability of intestinal Rotav and Colost 1981; Bohl nursing pi of the lac with trans U ilaltErmanr 27 crypts resulting in slower replacement of damaged aborptive epithelial cells in younger pigs (Moon, 1971). One or more of these explanations may apply to the age-resistance shown by rotavirus infection. The age-dependence of rotavirus infection may also reflect a difference in the number of virus—specific receptors on enterocytes of different maturity (Wolf et al, 1981). Riepenhoff-Talty et al (1982a) found fewer rotavirus receptors on enterocytes in the intestine of adult mice than in mice less than 11 days of age. However, this does not completely explain the difference in age susceptibility. Although mice greater than 14 days of age are refractory to clinical disease, they are infected, and the extent and distribution of antigen is similar to that seen in 7-day—old mice with rotaviral diarrhea (Eydelloth et al, 1984). Thus, the ability of mice to resist disease is not simply related to the ability of the virus to infect certain portions of the intestinal tract. Rotavirus antibody is present in the serum, milk, and colostrum of most, if not all, adult sows (Corthier, 1981; Bohl et a1, 1984). Thus the clinical signs in nursing piglets will be influenced by the immune status of the lactating sow, a situation similar to that found with transmissible gastroenteritis (Hooper and Haltermann, 1966). As expected, removal of nursing piglets from the influence of "lactogenic" immunity, such as occurs a (Woode and Serum rotaviral d (Mouwen et al, l977). children hc found isotc acidosis, e the specif: also was re study of 2' caused by 1 acidosis a (Tallet et Of these c hypernatre Serum then increased These labc vomiting 6 illness it Some Piglets We as the Cam eff ‘veCt Of 28 as occurs at weaning, increases the severity of diarrhea (Woode and Bohl, 1981). Clinical Pathology Serum biochemical alterations associated with rotaviral diarrhea have not been studied in great detail (Mouwen et al, 1972; Tallett et al, 1977; Rodriguez et a1, 1977). Rodriguez et a1 (1977), in a study of 72 children hospitalized with rotavirus gastroenteritis, found isotonic dehydration, compensated metabolic acidosis, and elevations in blood urea nitrogen (BUN) and the specific gravity of urine. Hypertonic dehydration also was reported, although much less commonly. In a study of 27 children hospitalized with acute diarrhea caused by rotavirus, most had mild compensated metabolic acidosis associated with decreased plasma bicarbonate (Tallet et al, 1977). Serum sodium concentration in most of these children was normal, but in 6 instances mild hypernatremia was detected. Additional alterations in serum chemistry included mildly decreased potassium, increased chloride, and elevated, normal, or reduced BUN. These laboratory findings reflect the high frequency of vomiting and dehydration associated with rotaviral illness in people. Some of the biochemical aspects of "white scours" in piglets were investigated before rotavirus was recognized as the cause of this condition (Mouwen et al, 1972). The effect of "white scours” on serum lipids indicated increased n6 phospholipic alterations The pa' but more se‘ serum bioch studied mor Cornelius e 2-day-old p that became and total I fluids ass< et al, l98. protein se (Cornelius those repo serum bioc Ectabolic hypoglycen (Cornelius a1 (1984) that may 1 mOrtality 0r Older from 6 to rOtaviral \7 29 increased neutral fat and decreased total cholesterol and phospholipid. These investigators noted that serum lipid alterations resembled those found in sprue in people. The pathophysiology of TGE of swine is similar to, but more severe than that of rotaviral enteritis. The serum biochemical changes associated with TGE have been studied more extensively (Reber and Whitehair, 1955; Cornelius et a1, 1968; Drolet et al, 1984). When 2-day-old piglets were challenged with TGE virus, those that became moribund had increased packed-cell—volumes and total proteins which were attributed to the loss of fluids associated with vomiting and dehydration (Drolet et a1, 1984). Elevations in packed—cell—volume and total protein secondary to TGE have been reported by others (Cornelius et al, 1968), but are in disagreement with those reported by Reber and Whitehair (1955). Additional serum biochemical changes associated with TGE were metabolic acidosis, increased BUN, elevated chloride, hypoglycemia, and unchanged sodium and potassium (Cornelius et al, 1968; Drolet et al, 1984). Drolet et al (1984) considered hypoglycemia a major feature of TGE that may be responsible for the high rate of neonatal mortality with that disease. Piglets 5 to 6 days of age or older are refractory to hypoglycemia even if fasted from 6 to 7 days (Sampson et al, 1942). Because rotaviral diarrhea is less common than TGE in piglets younger than 7 days of age, hypoglycemia may not be as important it Rotavi' the Villous membrane-b0 sodium-pota al, l984). accompaniec decreased e l984). The 3. activities the strain example, D lO-day-olc had decree “PPEI jEjl infected 1 sodium-p0. ileum Onl- Loss to nutrie with Porc Stool Osn m0STU/lite Of the o: l 30 important in rotaviral infections. Pathophysiology Rotavirus infection in pigs causes desquamation of the Villous epithelial cells resulting in loss of membrane-bound enzymes such as lactose, sucrose, and sodium-potassium ATPase (Davidson et al, 1977; Graham et al, 1984). Loss of microvillus-associated enzymes is accompanied by reduced transport of 3-0—methyglucose and decreased absorption of sodium and water (Graham et al, 1984). The segments of intestine in which altered enzyme activities are detected may, in part, be influenced by the strain of rotavirus used to infect the piglets. For example, Davidson et a1 (1977) found that 8- to lO-day-old piglets infected with a human rotavirus strain had decreased sodium-potassium ATPase activity in the upper jejunum. In contrast, when Graham et a1 (1984) infected piglets with a porcine rotavirus (OSU strain), sodium-potassium ATPase activities were depressed in the ileum only. Loss of membrane—associated digestive enzymes leads to nutrient malabsorption. Miniature piglets infected with porcine rotavirus (OSU strain) had an increase in stool osmolarity from 248 i 20 mOsm/liter to 348 i 20 mOsm/liter at 75 hours after inoculation. The majority of the osmotic gap in the feces could be accounted for by lactose in the stools. Sodium concentration in the stool increased, b1 1984). Male! uptake of D- rotavirus-in al, l977;‘Wc These 5 events in tl rotavirus d mucosal sur reduced, an malabsorpti At ne rotavirus stomachs o Partially tine betwe and Under( refleCtS . Small int Cecum and in lactea rOtaVlrus inoculat: atrophy . JerUUm, 31 increased, but potassium stayed the same (Graham et al, 1984). Malabsorption also has been measured by a reduced uptake of D-xylose in the small intestine of rotavirus-infected children and calves (Mavromichaelis et al, 1977; Woode et al, 1978). These studies indicate the following sequence of events in the pathophysiology of rotaviral diarrhea: (a) rotavirus destroys columnar Villous epithelial cells, (b) mucosal surface area and important digestive enzymes are reduced, and (c) osmotic diarrhea occurs due to nutrient malabsorption. Lesions and Pathogenesis Gross Lesions At necropsy, lesions in piglets infected with rotavirus are confined to the intestinal tract. The stomachs of experimentally—infected piglets are usually partially filled with milk regardless of differences in time between the last feeding and necropsy (Torres-Medina and Underdahl, 1980). The color of the fecal material reflects the diet and may be white, yellow, or grey. The small intestinal wall is reduced in thickness, and the cecum and colon may be distended with fluid. Visible fat in lacteals in gnotobiotic piglets infected with rotavirus is absent between 16 and 72 hours after inoculation (Theil et al, 1978). Evidence of Villous atrophy in the small intestine, especially in the jejunum, can be seen by subgross examination (Pearson and ltNulty, 197 Histopatholc The hi: are similar al, l97lb; ' et al, 1983 shortening epithelial and exposur quickly re' with no mi biochemica 1978). Diarr Small inte within 1- Villous a after the (BOhl et hours aft some gas, Crouch a7 to 168 h The at whict and Thej early sw 32 McNulty, 1977). Histopathologic Lesions The histopathologic lesions of rotavirus infection are similar in many mammalian and avian species (Mebus et al, 1971b; Theil et al, 1978; Conner et a1, 1980; Johnson et a1, 1983). The lesions are characterized by shortening of small intestinal villi, desquamation of epithelial cells lining the distal portion of the villus, and exposure of the lamina propria. The latter is quickly relined by squamous to cuboidal epithelial cells with no microvillous border (Woode et al, 1976b) and with biochemical characteristics of crypt cells (Middelton, 1978). Diarrhea in piglets precedes the development of small intestinal lesions. If small intestine is obtained within 1-3 hours after the onset of diarrhea, little Villous atrophy is evident, but if greater than 12 hours after the onset of diarrhea, Villous atrophy is distinct (Bohl et a1, 1978). Villous atrophy is most severe 24-72 hours after infection; villi may be essentially absent in some cases (Pearson and McNulty, 1977; Theil et al, 1978; Crouch and Woode, 1978). Villous atrophy persists for 48 to 168 hours after infection (Pearson and McNulty, 1977). There is disagreement about the stage of infection at which fusion of villi occurs. McAdaragh et a1 (1980) and Theil et a1 (1978) reported fusion of villi in the early stages of infection whereas Pearson and McNulty (1977) and T fusion of vi The re} lesions in 1 variable. atrophy at jejunum, an villous atr small intes killed 36 c severe vili investigat and distal duodenum ( Torres-Med In one of Piglets, t duodenal r the pigs lesions 0 intestine gnotobiot middle sn in the up Diffeteh( methods ‘ 33 (1977) and Torres—Medina and Underdahl (1980) noticed fusion of villi only after several days. The reported distribution of small intestinal lesions in piglets infected with porcine rotavirus is variable. Pearson and McNulty (1977) found villous atrophy at all levels of the intestine (duodenum, jejunum, and ileum). McAdaragh et a1 (1980) also noticed villous atrophy and fusion of villi at all levels of the small intestine. In fact, the duodenum of 2 piglets killed 36 or 72 hours after infection, respectively, had severe villous atrophy and fusion of villi. Other investigators have found lesions primarily in the middle and distal small intestine and "less strikingly" in the duodenum (Woode et al, 1976; Crouch and Woode, 1978; Torres-Medina and Underdahl, 1980; Graham et al, 1984). In one of the earliest reports of "white scours" in piglets, the distribution of lesions varied. The duodenal mucosa was most severely altered in one—third of the pigs (Mouwen et a1, 1971). One-half of the pigs had lesions of greatest severity in the distal small intestine. When a bovine rotavirus strain was given to gnotobiotic piglets, lesions were most severe in the middle small intestine, but milder lesions were detected in the upper small intestine (Hall et a1, 1976). Differences in age of the piglets, virus strains, and methods of sampling the intestine might explain these differences in lesion distribution. Glandul epithelial < increased p1 gnotobiotic and is maxi et al, l980 porcine rot and mid-sme al, l984). and ileum ( strain of ‘ (McAdaragh (1983) not gnotobioti preceded t The s is influer Et a1 (19. Virus (gr and mild ePithelia Villous 5 et a1 (1‘. aSSOCiaU (ch 1) a- Specific 34 Glandular crypts respond to the loss of villous epithelial cells caused by rotavirus infections by increased proliferation. Crypt hyperplasia in gnotobiotic piglets occurs by 48—60 hours after infection and is maximal by 96 hours (Theil et al, 1978; McAdaragh et a1, 1980). Piglets infected with the OSU strain of porcine rotavirus had crypt hyperplasia in the jejunum and mid-small intestine but not in the ileum (Graham et al, 1984). However, crypt hyperplasia in the duodenum and ileum of gnotobiotic piglets infected with a field strain of porcine rotavirus has been reported by others (McAdaragh et al, 1980). Interestingly, Johnson et a1 (1983) noted that crypt hyperplasia in the duodenum of gnotobiotic pups infected with canine rotavirus was not preceded by villous atrophy. The severity and distribution of intestinal lesions is influenced by the type of infecting rotavirus. Theil et a1 (1985) studied the pathogeneis of a rotavirus-like virus (group B) in gnotobiotic piglets and found variable and mild histologic changes. Sloughing of villous epithelial cells occurred only at the tips of villi and villous atrophy was evident only occasionally. McNulty et al (1983) compared the clinical and virologic findings associated with infection by a group A avian rotavirus (ch 1) and a non—group A avian rotavirus (ch 132) in specific—pathogen—free chickens. There was a distinct difference between the two rotavirus isolates in viral antigen dis had a predi rotavirus s the mid-sma difference: of rotavirr Eosin' observed h mice infec 1967), rat (Vonderfec al, l98la) only with inclusion: correlate structure The - unCOmplic gastric m and McNul Viral ant 1977; The deSCribe( epitheli; rotaViru. CEcum an mlCrOsCO 35 antigen distribution. Rotavirus strain ch 1 (group A) had a predilection for the duodenal mucosa, whereas rotavirus strain ch 132 (non—group A) replicated best in the mid-small intestine. This was the first report of differences in intestinal tropism for different isolates of rotavirus. Eosinophilic cytoplasmic inclusions have been observed histologically in the intestinal epithelium of mice infected with murine rotavirus (Adams and Kraft, 1967), rats infected with an atypical rotavirus (Vonderfecht et al, 1984), and in cell culture (Carpio et al, 1981a). Inclusions, in most instances, were evident only with the use of special stains. The nature of the inclusions is unclear. Adams and Kraft (1967) could not correlate inclusions seen by light microscopy with structures seen by electron microscopy. The pathogenesis of rotavirus infections tends to be uncomplicated by lesions in other organs. Lesions in the gastric mucosa have been reported (Woode 1975b; Pearson and McNulty, 1977) but have not been associated with viral antigen (McNulty et al, 1976a; Davidson et al, 1977; Theil et a1, 1978). Rodriguez et a1 (1980) described cecal and colonic vacuolation of superficial epithelial cells in mice infected with a murine rotavirus. Murine rotavirus has been detected in the cecum and colon of mice by immunofluorescent and electron microscopic techniques (Adams and Kraft, 1967; Narita et al, 1982). Scanning Ele Scannir offers the a specimens, : visualized l advantages, studied by Torres-Hedi Underdahl ( the intesti given a rot case of p01 Visualized lower smal occluded t 36 hours a lensth of Cells expc epithelial epithelia] dearly n01 There is < fuSion 0c. quiOn as Edged V11 Underdahl 36 al, 1982). Scanning Electron Microscopy Scanning electron microscopy (SEM) of intestine offers the advantages of tridimensional view, larger specimens, and observation of details not readily visualized by histologic examination. Despite these advantages, porcine rotavirus infections have been studied by SEM infrequently (McAdaragh et a1, 1980; Torres-Medina and Underdahl, 1980). Torres-Medina and Underdahl (1980) described the sequential SEM changes in the intestinal mucosa of 6—day—old gnotobiotic piglets given a rotavirus strain (B—3l7) isolated from a field case of porcine rotaviral diarrhea. Lesions were first visualized at 12 hours after infection in the middle and lower small intestine. Enterocytes were swollen and occluded transverse furrows and goblet cell openings. By 36 hours after inoculation, villi were one—third the length of control villi and detachment of epithelial cells exposed lamina propria. Microvilli on degenerate epithelial cells were sparse and short. Regeneration of epithelial cells was evident by 156 hours and villi were nearly normal in dimension by 16 days after infection. There is disagreement about the time at which villous fusion occurs. McAdaragh et al (1980) observed villous fusion as early as 12 hours post—inoculation (PI), and fused villi were numerous by 72 hours. Torres—Medina and Underdahl (1980) did not observe fused villi until 4.8 days PI. Transmissic Rotavi electron mi piglets by Virions at within ves: The earlie; between l8 swelling, cytoplasmi distended endoplasmi Cells with be seen. synthesize Visible it Narita St The j Observed } after rot. Piglets i in the mi (PEarson Et al, 19 37 days PI. Transmission Electron Microscopy Rotavirus particles can be seen by transmission electron microscopy (TEM) in the jejunum of gnotobiotic piglets by 12 hours after infection (Narita et al, 1982). Virions at this time are on and between microvilli and within vesicles in the apical cytoplasm and terminal web. The earliest cytopathologic alterations, detectable between 18-24 hours after infection, are cytoplasmic swelling, reduction in the number and size of microvilli, cytoplasmic lipid droplets, rough endoplasmic reticulum distended with vacuoles, and virus particles in the rough endoplasmic reticulum. By 48 hours, immature cuboidal cells with an uneven, incomplete microvillous border can be seen. No virus particles are evident in newly synthesized cells. Virus particles may or may not be visible in goblet cells (Pearson and McNulty, 1979; Narita et al, 1982). Immunofluorescence and Location of Viral Antigens The location and number of antigen—positive cells observed by immunofluorecence (IF) is a function of time after rotavirus infection. Maximal fluorescence in piglets is observed in the enterocytes near villous tips in the middle small intestine 24 hours after infection (Pearson and McNulty, 1977; Crouch and Woode, 1978; Bohl et al, 1978). McAdaragh et a1 (1980) first observed fluorescence in the duodenum and upper jejunum of gnotobiotic fluorescenc ileum by 36 with findir 1977; Bohl and Underda infected v: intestine, ileum. Du not observ rotaviruse for 96 hou et al, 197 infected v than 24 h< Viraf than smal seen in t' Piglets ( and calve nodes of a1, l97lt intesting SPECifiC Were Pre: PIESehCe prOPI‘ia ' 38 gnotobiotic piglets 12 hours after infection. The fluorescence progressively descended through the upper ileum by 36 hours. These findings are in disagreement with findings by other investigators (Chasey and Lucas, 1977; Bohl et a1, 1978; Theil et al, 1978; Torres—Medina and Underdahl, 1980). The latter investigators found infected villous epithelial cells throughout the small intestine, but most consistently in the jejunum and ileum. Duodenal fluorescence was sparse and variable or not observed. Enterocytes infected by group A rotaviruses desquamate so IF is only detected in piglets for 96 hours (McAdaragh et al, 1980) to 168 hours (Theil et a1, 1978) after infection. Gnotobiotic piglets infected with a group B rotavirus had few IF cells later than 24 hours after infection (Theil et al, 1985). Viral antigen has been detected by IF in sites other than small intestine epithelium. Immunofluorescence was seen in the lamina propria of the small intestine of piglets (Theil et al, 1978), dogs (Johnson et al, 1983), and calves (Mebus et al, 1971b), and the mesenteric lymph nodes of piglets and calves (Theil et a1, 1978; Mebus et al, 1971b). Fluorescence observed in the small intestinal lamina propria of calves was not regarded as specific by Mebus et al (1971b) because many eosinophils were present. But Stair et a1 (1983) substantiated the presence of rotavirus in macrophages in the lamina propria by TEM. A few infected enterocytes have been detected in (Narita et ; (Banfield e l983), and 1985). Compar negatively time. Elev content fr signs cont are nunero greatest r from pigs 1976; Bob? Particles 5‘8 days TOrres-Me Resi neutrali; tract (P. 1978; Of secreted infectic paSSiVel 39 detected in the cecum of rotavirus—infected piglets (Narita et a1, 1982), the colon and rectum of mice (Banfield et a1, 1968), colon of chickens (McNulty et al, 1983), and cecum and colon of calves (Reynolds et a1, 1985). Viral Shedding Compared to IF, rotavirus can be detected in negatively stained fecal material for a longer period of time. Electron microscopic examination of intestinal content from piglets just beginning to show clinical signs contain few rotaviral particles. Viral particles are numerous by 24 hours after infection, and the greatest number of particles are evident in specimens from pigs 48 to 72 hours after infection (Lecce et al, 1976; Bohl et al, 1978). The number of rotavirus particles is reduced or they cannot be detected between 5-8 days after infection (Tzipori and Williams, 1978; Torres-Medina and Underdahl, 1980). Law Humoral Immunity Resistance to rotavirus diarrhea is mediated by neutralizing antibodies present within the intestinal tract (Pearson and McNulty, 1977; Snodgrass and Wells, 1978; Offit and Clark, 1985). These antibodies may be secreted into the intestinal lumen following intestinal infection (Corthier and Vannier, 1983) or be obtained passively via colostrum and milk (Bridger and Brown, 1981; Saif antibody dc infection l985). Passi suckling p adequate a rotavirus lgG, lgA, Bachmann, predomina1 persisting class whi class in The Pigs less there are infrequer fEEd (Bot antibody weeks af' on SElec- an inCre at this Rot inteStin EECQS b> 40 1981; Saif et a1, 1984). The presence of serum rotavirus antibody does not correlate with resistance to rotavirus infection (Snodgrass and Wells, 1978; Offit and Clark, 1985). Passive immunity against enteric pathogens occurs in suckling pigs as a result of frequent ingestion of an adequate amount of specific antibodies. Antibodies to rotavirus in swine occur in the immunoglobulin classes IgG, IgA, and IgM in mammary secretions (Hess and Bachmann, 1981). The IgG class of immunoglobulin predominates in colostrum, but the highest and longest persisting rotavirus antibody is associated with the IgA class which constitutes the predominant immunoglobulin class in the milk of pigs (Vairman et a1, 1970). The antirotaviral antibody in sows' milk protects pigs less than 7-days-old from rotavirus infection unless there are inadequate antibody titers in the milk, infrequent suckling, or dilution of antibodies by creep feed (Bohl et al, 1978). Protective IgA antirotaviral antibody is found in sows' milk until weaning at 6 to 8 weeks after parturition (panel report on the Colloquium on Selected Diarrheal Diseases of the Young, 1978). Thus an increased incidence of rotavirus diarrhea is observed at this time. Rotavirus infection induces local production of intestinal antibody. Immune complexes appeared in the feces by 4 days after experimental inoculation of newborn piglets with were demonstr and Vannier, feces of hum: clinical sig1 (Stals et al evidence of \dth rotavir ammnestic r production c lived (42-7( same rotavi: infected (B The ex hOSt reSpop understood, some aSpect infection 1983), In imflmnitv a 2 days aft days after 9 SlmultaneC aneStigat 41 piglets with rotavirus. Free IgA and IgM coproantibodies were demonstrated by 7 days after inoculation (Corthier and Vannier, 1983). Maximal excretion of IgA in the feces of human infants occurred 7 days after the onset of clinical signs and corresponded with clinical improvement (Stals et al, 1984). In human infants, there also is evidence of an immunologic memory because reinfection with rotavirus is associated with an intestinal IgA anamnestic response (Yamaguchi et al, 1985). The production of intestinal antibody in calves may be short lived (42-70 days), thereby allowing reinfection with the same rotavirus strain with which they were originally infected (Bachmann et al, 1983). Cell-Mediated Immunity The exact role of the different components of the host response to rotavirus infection is not clearly understood. Attention has been given only recently to some aspects of the cellular immune response to rotavirus infection (Riepenhoff—Talty et al, 1982b; Little et a1, 1983). In Balb—c mice, virus—specific cell mediated immunity appeared in the splenic lymphocytes as early as 2 days after rotavirus infection and peaked around 10 days after infection (Riepenhoff—Talty et al, 1982). Because cell—mediated immunity (CMI) increased simultaneously with cessation of viral replication, these investigators concluded that CMI may be important in recovery from the disease. Littl lymphocyte Infant mud and T lymp receiving animals re that did 1 important Rota by Eiden experienc which was immunocon seronegat similar e both the resolved antibody dEEense fUnction antibody Cel transfei IeUkOCYT i985), PaSsive prOtth 42 Little et al (1983) studied the role of T and B lymphocytes in recovery from rotaviral infections. Infant nude mice were transfused with equal numbers of B and T lymphocytes from syngenic adult mice. Animals receiving spleen cells from immunologically mature animals resolved their infections more quickly than those that did not, which suggested that T and B cells were important in the resolution of the rotavirus infection. Rotavirus infection in athymic nude mice was studied by Eiden et a1 (1986). Neonatal T-cell-deficient mice experienced a self—limited gastrointestinal infection which was identical to that observed in age-matched immunocompetent mice. Also, adult T—cell-deficient seronegative mice and age—matched normal mice showed a similar extent of resistance to rotavirus infection. In both the neonatal and adult mice, the infection was resolved without the generation of antirotavirus antibody. These investigators suggested that the host defense against murine rotavirus requires neither functional T—lymphocytes nor specific antirotaviral antibody. Cell—mediated immunity to rotavirus can be passively transferred to neonatal calves by giving colostral leukocytes from immunized heifers (Duhamel and Osburn, 1985). Although significant cell-mediated immunity is passively acquired by the calves, it does not confer protection when calves are orally challenged with live bovine rotav The big gastroenter: underscores Commercial rotavirus s disappointi 1986). Two a] rotavirus stinulatio newborn ca modified 1 afforded p hours afte Furthermo neonatal (Thurber trials fa RadOStitg It 1 antibodi newborn Zaane et rotaVirr antibgdj 43 bovine rotavirus. Immunization The high morbidity and mortality caused by rotavirus gastroenteritis in many mammalian and avian species underscores the need for an efficacious and safe vaccine. Commercial vaccines containing bovine or porcine rotavirus strains are available, but results have been disappointing (Hoblet et a1, 1984; Van Zaame et a1, 1986). Two approaches have been used to try to prevent rotavirus infections in calves. The first involves stimulation of active immunity in the intestinal tract of newborn calves by oral inoculation with attenuated modified live rotavirus vaccines. These vaccines afforded protection to gnotobiotic calves as early as 48 hours after inoculation (Mebus et a1, 1973). Furthermore, use of this vaccine was reported to decrease neonatal calf morbidity and mortality in field trials (Thurber et al, 1977). However, other double-blind trials failed to substantiate these findings (Acres and Radostits, 1976). It has been suggested that colostral rotavirus antibodies may interfere with active immunization of the newborn calf. This has been documented recently (Van Zaane et a1, 1986). Oral vaccination, with modified live rotavirus, of calves fed colostrum with rotavirus antibodies did not induce a protective intestinal immune response. immunity i1 antibodies delay or o: A see rotavirus with subse calf via c cows with increased and milk, lZ supple] experimen increase multiple 81, 1983) initially cows to 1 bovine r< (1984) h. will res With whi instance A u rbtavirt to nursj .1 u «lcense, 44 response. Oral vaccination did induce protective immunity in calves which were not fed colostral rotavirus antibodies, for practical purposes, it is not sensible to delay or omit colostrum feeding. A second approach used to protect calves from rotavirus infection is immunization of the pregnant dam with subsequent passive transfer of antibodies to the calf via colostrum and milk. Parenteral immunization of cows with an adjuvated modified live rotavirus vaccine increased virus—neutralizing antibody titers in colostrum and milk, which were protective when fed to calves as a 1% supplement (Saif et al, 1983). In the same experiment, a commercially available vaccine did not increase virus-neutralizing antibody titers. Because multiple serotypes of bovine rotavirus exist (Murakami et al, 1983) and do not show cross protection, it was initially believed that for parenteral immunization of cows to be effective, a vaccine must contain all of the bovine rotavirus serotypes. However, Snodgrass et a1 (1984) have shown that after monovalent vaccination, cows will respond heterotypically to all rotavirus serotypes with which they were exposed. Therefore, in some instances, single serotype vaccination may be sufficient. A modified-live TGE virus and serotype 1 porcine rotavirus vaccine to be administered to pregnant swine or to nursing piglets prior to weaning is federally licensed. There are no published controlled studies on the efficac: have shownii of diarrhea gains betwe l984). The calf vaccin this has be does not re does it prc Rotavf causes of 2 years of a; need for a: has been r develOpmen Strain as under acti a1, 1979). administre induces re moderate 1 rOtavirus. Anot adVantage genetic r CO-infeet “O“‘CUIti 45 the efficacy of this vaccine, but preliminary studies have shown no differences in the incidence or frequency of diarrhea, rotavirus shedding, and post-weaning weight gains between vaccinates and controls (Hoblet et al, 1984). The rotavirus strain in a commercially-available calf vaccine has been used to vaccinate swine. However, this has been shown to be ineffective because the virus does not replicate in newborn colostrum-free piglets nor does it protect them from disease (Lecce and King, 1979). Rotaviruses are now considered one of the major causes of gastroenteritis in children younger than two years of age (Flewett and Woode, 1978). Thus, an urgent need for an effective and safe vaccine for use in people has been recognized. A number of approaches to vaccine development are being studied. Use of a bovine rotavirus strain as a potential vaccine candidate for people is under active investigation (Zissis et a1, 1983, Wyatt et al, 1979). Vesikari et a1 (1983) have shown that oral administration of a bovine rotavirus (NCDV strain) induces resistance in infants and young children against moderate to severe diarrheal illness caused by human rotaviruses. Another approach to vaccine development takes advantage of the propensity of rotaviruses to undergo genetic reassortment (Greenberg et al, 1981). Co-infection of cell cultures with animal rotaviruses and non—cultivatable human rotaviruses has yielded reassortar the major and the al rotavirus type repr Ente other ent pigs. Al assist ir insuffici definitii the demOI diarrhei the pres Man been des sensitiv often he StOpa, ] The labc are GHZj micresc Mm En are sin utiliZc 46 reassortants which contain the gene segment coding for the major neutralization protein from the human rotavirus and the ability to replicate in yitgg from the animal rotavirus (Midthun et a1, 1985). Reassortants of this type represent potential live vaccine strains. Diagnosis Enteric disease caused by rotavirus and several other enteropathogens is common in nursing and weanling pigs. Although history, clinical, and necropsy findings assist in obtaining an accurate diagnosis, they alone are insufficient to distinguish among these diseases. A definitive diagnosis of rotavirus infection depends on the demonstration of rotaviral antigen in specimens from diarrheic animals and correlation of these findings with the presence of histopathologic lesions. Many useful techniques for rotavirus detection have been described, and the details of the relative sensitivities, specificities, and advantages of each test often have been provided (Yolken et a1, l978a; Yolken and Stopa, 1979; Benfield et a1, 1984; Reynolds et al, 1984). The laboratory tests that have been most widely compared are enzyme—linked immunosorbent assays (ELISA), electron microscopy (EM), and immunofluorescence (IF). Enzyme—linked Immunosorbent Assays Enzyme—linked immunosorbent assays are systems that are similar in design to radioimmunoassays, but which utilize an enzyme rather than radioactive isotope as the immunoglol enzyme, wd antigen-a1 substrate color, ca: colorimet determine that can a modific was used modificat detectior ELISA or The rotaviru; were mor. ReYnolds several of the E microscc l984). Coo for the 1984; Re Compare. detecri ResUlts 47 immunoglobulin marker (Engvall and Perlmann, 1972). This enzyme, when bound to the "solid phase" in a series of antigen-antibody reactions, interacts with added substrate to produce a product. The product, usually a color, can be detected visually or with the aid of a colorimeter. The sensitivity of these assays is determined by the lowest concentration of visible color that can be detected. Yolken and Stopa (1979) described a modification of a rotavirus ELISA in which a substrate was used that yielded a fluorescent product. This modification improved the sensitivity of human rotavirus detection by greater than 100 times compared to standard ELISA or radioimmunoassay techniques. The sensitivity of ELISA has been compared to other rotavirus detection methods. In most instances, ELISA were more sensitive than EM (Rubenstein and Mille, 1982; Reynolds et al, 1984). For example, comparison of several commercially—available ELISA indicated that one of the ELISA was more sensitive (98%) than immunoelectron microscopy (93%), IF (86%), and EM (842) (Mounet et a1, 1984). Commercially—available ELISA correlate well with EM for the detection of bovine rotavirus (Benfield et al, 1984; Reynolds et al, 1984). Reynolds et a1 (1984) compared EM to enzyme—linked immunosorbent assay for the detection of rotavirus and coronavirus in bovine feces. Results showed excellent correlation with the detection of rotavir calves as Benfield t immunosorl than IF 0: rotavirus enzyme-lit detecting enzyme-li is not se rotavirus other lab An a viral ant for detec (1977) he feces is DisadVant et al, 19 Viral ant feces of I984), M Bec. animals f remains (I978b) 48 of rotavirus by EM in feces from experimentally infected calves as well as from diagnostic laboratory specimens. Benfield et a1 (1984) found a commercial enzyme-linked immunosorbent assay as sensitive as EM but more sensitive than IF or viral isolation for detecting bovine rotavirus. However, EM was more sensitive than IF, enzyme-linked immunosorbent assay, or viral isolation for detecting porcine rotavirus. The commercially—available enzyme-linked immunosorbent assay used by Benfield et a1 is not sensitive for the detection of porcine rotaviruses, so it should be used in combination with other laboratory procedures. An advantage of ELISA when compared to EM is that viral antigen need not be assembled into viral particles for detection. This is useful because Mathan et a1 (1977) have shown that much of the rotavirus antigen in feces is not assembled into viral particles. Disadvantages of some ELISA are non—specificity (Chrystie et al, 1983) and in some instances, inability to detect viral antigen-antibody complexes which commonly occur in feces of naturally infected animals (Crouch and Acres, 1984). Electron Microscopy Because many rotaviruses associated with diarrhea in animals have been difficult to adapt to cell culture, EM remains the primary method for their detection. Flewett (l978b) reported that, with skill and perseverance, EM will detect should not rapidly die improved, 1 particles This techn The sensit use of sol technique antirotavf microscopj direct EM et al, 19 All that can and comp] group spr the grou‘ group A of non~g the COmn inn against rOtavirr these I, 49 will detect 105 viral particles/ml, 106 particles/ml should not be missed, and 108 particles/ml will be rapidly diagnosed. These sensitivities can be greatly improved, however, by the aggregation of rotavirus particles with antisera prior to EM (Saif et a1, 1977). This technique is termed immunoelectron microscopy (IEM). The sensitivity of EM also can be greatly enhanced by the use of solid—phase immunoelectron microscopy. This technique utilizes protein coated grids treated with antirotavirus antibody. Solid phase immunoelectron microscopy has been reported 30 times more sensitive than direct EM and 10 times more sensitive than ELISA (Yolken et al, 1979). Immunofluorescence All group A rotaviruses possess a common antigen that can be detected by IF, IEM, ELISA, gel diffusion, and complement fixation (Woode et al, 1976a). Thus, group specific antigen and antisera prepared from any of the group A rotaviruses can be used for the diagnosis of group A rotavirus infections in any species. Detection of non-group A rotaviruses requires antisera specific for the common antigen of each group. Immunofluorescence techniques which utilize antisera against the common antigen of group A porcine rotaviruses have been described (Theil et al, 1978). In these reports, it has been demonstrated that rotaviral antigen in intestinal smears or frozen sections can usually 0T Thus, in i must be s< infection disadvant. interpret fluoresce decomposi Other Dia Othe describec Woode an< gnotobior presence negative the most rotaviru At Preventj Vaccines Van ZaaI haVG as of thes Th and mm 50 usually only be detected for 72-96 hours after infection. Thus, in field cases of rotaviral diarrhea, specimens must be selected from piglets in the early stages of infection if IF tests are to be successful. Additional disadvantages of IF procedures are subjective interpretation, interference by non-specific background fluorescence, and interference by post mortem decomposition. Other Diagnostic Techniques Other techniques for rotavirus detection have been described (Middleton et al, 1974; Middleton et al,l976; Woode and Bohl, 1981). One of these, the inoculation of gnotobiotic piglets, has been utilized to demonstrate the presence of rotavirus when other techniques have proved negative. Inoculation of the gnotobiotic piglet may be the most sensitive means of demonstrating the presence of rotavirus (Woode and Bohl, 1981). Treatment and Control At present, there are no practical methods for preventing rotavirus infection; commercially available vaccines have not proved efficacious (Hoblet et al, 1984; Van Zaane et al, 1986). Management factors, therefore, have assumed an important role in moderating the severity of these infections. Thorough cleaning and disinfection of the farrowing and nursery units and the use of "all—in-all-out" management systems will reduce the amount of virus in the piglets' to which delay the 1980). R environme cannot be (Snodgras Unde ambient t associate and Woode piglets, piglets 1 caused b Res neutrali tract (S Most sow antibodj immunity and Bacl Way is : rotavir. antibod- and Boh at birt SeVerit 51 piglets' environment. Reduction of the quantity of virus to which piglets are exposed may reduce the severity and delay the onset of rotaviral diarrhea (Lecce and King, 1980). Rotaviruses are extremely resistant to environmental conditions and disinfectants, so they cannot be completely eliminated from the farm environment (Snodgrass et a1, 1977; Sattar et a1, 1983). Under natural and experimental conditions, a drop in ambient temperature has been shown to increase mortality associated with rotaviral infection in piglets (Crouch and Woode, 1978). Meeting ambient temperature needs of piglets, especially preventing chilling of neonates and piglets with diarrhea, assists in controlling losses caused by rotavirus. Resistance to rotaviral diarrhea is mediated by neutralizing antibodies present within the intestinal tract (Snodgrass and Wells, 1978; Offit and Clark, 1985). Most sows are positive for group A rotavirus serum antibodies and will transfer variable amounts of passive immunity to their piglets via colostrum and milk (Hess and Bachmann, 1981). Passive protection obtained in this way is important because a low infectious dose of rotavirus in the presence of protective milk—secreted antibody may result in immunity without disease (Woode and Bohl, 1981). If piglets receive rotavirus antibodies at birth, the onset of infection will be delayed, and the severity of diarrhea and mortality caused by rotavirus will be r infection 2-weeks-o In g controlle contamine receive a warm, drj 52 will be reduced. This is important because rotavirus infections are most severe in piglets less than 2-weeks-old (Lecce et a1, 1976). In general, rotaviral diarrhea of pigs can best be controlled by avoiding environments that are heavily contaminated with rotavirus and by ensuring that piglets receive an adequate amount of colostrum and are kept warm, dry, and well fed. Six hysterot as descr Control inoculat pigs wer 25-30°C. York, NY on the f increase Miniats bEfore n Inn swabs fr residual each pie Cultured tergito] aerobic contamiI examiner MATERIALS AND METHODS Gnotobiotic Pigs Sixty-three pigs were obtained from 7 sows by closed hysterotomy and maintained in stainless steel isolators as described previously (Miniats and Joel, 1978). Control pigs were kept in isolators separate from inoculated pigs. Isolators containing these gnotobiotic pigs were kept in a room with an ambient temperature of 25—30°C. The pigs were fed 50 m1 of SPF-lac (Borden, New York, NY) three times (morning, afternoon, and evening) on the first day. The volume of milk replacer was increased by 13.5 ml/feeding daily as recommended by Miniats and Joel (1978). Pigs were fasted 3 to 6 hours before necropsy. Immediately prior to inoculation, fecal and oral swabs from 1 pig in each isolator and a composite swab of residual milk in feed pans were collected. Ileum from each pig was collected at necropsy. Swabs and ilea were cultured for bacteria by use of sheep blood, tergitol—seven, and brilliant-green agars incubated in aerobic and anaerobic atmospheres. Evidence of bacterial contamination also was obtained by histopathologic examination of intestine. 53 'Pe Micr¢ before in! cultured with the enteric o inoculati Bacteria intestine with rote necropsi. Viabilit by aerob Por Universj (Ohio Ag (OARDC) TOtavir‘ (Bohl e morphol Strain al, 197 intestj Percin, State . DaVId SD). 54 Microorganisms were not detected in any isolators before inoculation of the pigs. Bacillus sp. was cultured from the ileum of l pig 3 days after inoculation with the SDSU strain of porcine rotavirus. A few enteric organisms were cultured from 1 pig 7 days after inoculation with the OSU strain of porcine rotavirus. Bacteria were seen by histologic examination in the large intestine of 2 pigs necropsied 1 day after inoculation with rotavirus (l pig--OSU; l pig--SDSU) and 2 pigs necropsied 3 days after inoculation with the OSU strain. Viability of the observed bacteria was not demonstrated by aerobic and anaerobic culture. Kine-a Porcine rotavirus designated as the Ohio State University (OSU) isolate was obtained from Dr. Linda Saif (Ohio Agricultural Research and Development Center (OARDC), Wooster, OH). The OSU strain of porcine rotavirus has serotype 1 and subgroup I specificities (Bohl et a1, 1984; Hoshino et a1, 1984). Additional morphologic and antigenic properties of this rotavirus strain have been described (Saif et a1, 1977; Theil et a1, 1978). The other porcine rotavirus was from an intestinal homogenate of a pig with diarrhea. This porcine rotavirus was designated as the South Dakota State University (SDSU) isolate and was obtained from Dr. David Benfield (South Dakota State University, Brookings, SD). The SDSU strain of porcine rotavirus has not been isolated strain n (Gottfri (persona Oh), so rotaviru times in inoculat gnotobic contents respectf inocular collectv differei tract a' Prepare New Yor twice t increas intesti 10,000 Pipette ~70°c. by fiii (Millii (APD) . filtra 55 isolated in cell culture. Antisera against the SDSU strain neutralized a serotype 2 porcine rotavirus (Gottfried strain) at a titer of greater than 1:1024 (personal communication: Dr. Linda Saif, OARDC, Wooster, OH), so this strain was considered a serotype 2 porcine rotavirus. Each strain of virus was serially passaged 4 times in gnotobiotic pigs; diarrhea was observed in inoculated pigs at each passage. At the fourth gnotobiotic pig passage, the intestinal tract and contents from 2 pigs given the OSU or SDSU isolates, respectively, were aseptically collected 48 hours after inoculation. To avoid cross-contamination, intestine was collected and processed on different days and in different locations. A 50% suspension of intestinal tract and contents in Hanks' balanced salt solution was prepared by homogenization in a blender (Waring Blender, New York, NY). The homogenate was frozen and thawed twice to rupture infected epithelial cells, thereby increasing the quantity of released virus. The intestinal homogenate was centrifuged 20 minutes at 10,000 x g. Aliquots (10 m1) of supernatant were pipetted into sterile plastic tubes and refrigerated at -70°C. As needed, supernatant was thawed and sterilized by filtration through a series of Millipore filters (Millipore Corp., Bedford, MA) with average pore diameter (APD) of 0.65 pm, 0.45 mm, and 0.22 pm. Sterility of the filtrate was confirmed by bacterial culture as described above. 1 examined containe Feces) r were obs The adapted availabi virus p; and OSU gnotobi Tw Pigs, w virus p 10'6, 1 were p1 of pig: Viruse Piss < totavi with 1 Pools. Signs micro, Pigle in th 56 above. Negatively stained specimens of inocula were examined by EM. Filtrates of the SDSU and OSU isolates contained +4 (see Electron Microscopic Examination of Feces) rotavirus particles. No contaminating organisms were observed. Virus Infectivity Titrations The SDSU strain of porcine rotavirus has not been adapted to cell culture, so an in yitgg method was not available to determine accurately the number of infective virus particles in the viral pools. Therefore, the SDSU and OSU virus pools were titrated for infectivity in gnotobiotic pigs. Two litters, one containing 11 and the other 12 pigs, were used for the titration of the OSU and SDSU 5 9 virus pools, respectively. Various dilutions (10‘ 10'6, 10‘7, and 10_8) of the SDSU and OSU virus pools were prepared in Hanks' balanced salt solution. Groups of pigs inoculated with different dilutions and different viruses were housed in separate isolators. Groups of 3 5 dilution of the osu pigs (except the group given the 10' rotavirus, which contained 2 pigs) were inoculated orally with 1 m1 of either the diluted SDSU or OSU rotavirus pools. Pigs were examined three times daily for clinical signs of diarrhea. Feces were collected for electron microscopic examination 48 hours after inoculation. Piglets that became diarrheic and had rotavirus particles in their feces at 48 hours after inoculation were considers caused d: pig-infe for the 10-5.8’ For groups t age, 28 contain: rotaviru rotavir' pig was appropr Pigs) w solutic in eacl Pigs we Four p: SDSU, inocul Signs intern SUSPQI 57 considered infected. The dilution of the virus pool that caused diarrhea in 502 of the piglets was defined as one pig-infectious—dose fifty (PIDSO). The calculated PID50 -7.2 for the SDSU and OSU rotavirus pools were 10 and -5.8 10 , respectively. Experimental Design Forty gnotobiotic pigs were assigned to treatment groups by use of a random number table. At 3 days of age, 28 pigs were each inoculated orally with 1 ml containing 105 PID50 of either the OSU strain of porcine 1 rotavirus (14 pigs) or the SDSU strain of porcine rotavirus (14 pigs). Immediately after inoculation, each pig was fed SPF-lac to which an additional 1 m1 of the appropriate viral inoculum had been added. Controls (12 pigs) were inoculated with filtered Hanks' balanced salt solution in the same manner as the principals. Five pigs in each of the virus-inoculated groups and four control pigs were killed at 24 and 168 hours after inoculation. Four pigs in each of the three experimental groups (OSU, SDSU, and control) were killed at 72 hours after inoculation. Clinical Signs and Body Weight Control and infected pigs were observed for clinical signs 3 times daily at the time of feeding. Body weight was measured prior to inoculation and at 24 hour intervals thereafter. Body weights were measured by suspending pigs in a sling attached to a spring scale (Ohaus, F were fed Bloc vacutaine for the e and for ethylene Packed-c microhen profiles Medicine reagent: Industr Pi bYani barbite the 8m: micros. Scanni Was re last P appro; The re 1/3, flexu 58 (Ohaus, Florham Park, NJ) with accuracy to i20 gms. Pigs were fed after body weights were recorded. Clinical Pathology Blood collected at necropsy was dispensed into vacutainer tubes (Becton—Dikinson, Rutherford, NJ) which for the collection of serum contained no anti-coagulant and for the collection of whole blood contained ethylenediamine—tetracetic acid (EDTA). Packed—cell-volume of unclotted blood was measured by the microhematocrit method (Benjamin, 1978). Serum chemistry profiles were measured by the Laboratory of Clinical Medicine (Sioux Falls, SD). Technicon SRA 2000 and reagents for the "basic panel” from Technicon (Technicon Industrial Systems, Tarrytown, NY) were used. Necropsy Pigs were bled by cardiac puncture and then killed by an intracardiac injection of a solution containing barbiturates. Segments (3 cm in length) from 6 sites in the small intestine were removed immediately for light microscopy (LM), fluorescent antibody testing (FAT), and scanning electron microscopy (SEM). The first segment was removed at the duodenal—jejunal flexure (DU) and the last portion was removed from the lower ileum (LI) approximately 5 cm proximal to the ileocecal junction. The remaining segments were removed at approximately 1/6, 1/3, 1/2, and 2/3 the distance from the duodenal—jejunal flexure to the ileocecal junction and were designated as the uPPer intestine respecti‘ LM and S] A small intestin Karnovsk and refr was inci attachme straigh‘ formali rectum, In addi heart, neutral 335%! G of por serum Were h State Storee gamma modif (Plum Uhles 59 the upper jejunum (UJ), mid-portion of the small intestine (MI), lower jejunum (LJ), and upper ileum (U1), respectively. The lumen of each section of intestine for LM and SEM was rinsed with 10% neutral buffered formalin. A small portion (0.5 cm2) of each segment of small intestine, cecum, and colon was removed and immersed in Karnovsky's fixative at pH 7.4 (Karovsky, 1965) for SEM and refrigerated at 4°C. The remaining small intestine was incised longitudinally along the mesenteric attachments, pinned to rigid sections of cardboard for straight flat fixation, and immersed in neutral buffered formalin. Sections of unfixed stomach, cecum, colon, rectum, and mesenteric lymph node were collected for FAT. In addition to these tissues, lung, liver, kidney, brain, heart, thymus, pancreas, and spleen were immersed in neutral buffered formalin for LM. Preparation of Fluorescein-Conjugated Gamma Globulin Gnotobiotic pig hyperimmune serum to the SDSU strain of porcine rotavirus (serotype 1) and rabbit hyperimmune serum to the OSU strain of porcine rotavirus (serotype 2) were kindly supplied by Dr. David Benfield (South Dakota State University, Brookings, SD). Hyperimmune serum was stored at -70°C until used. Fluorescein-conjugated gamma-globulins were prepared from hyperimmune serum by a modification of procedures developed by Dr. I. C. Pan \ (Plum Island Research Laboratories, Plum Island, NY). Unless noted, all procedures were done at room temperat' flask. addition solutior magnetic been ade Precipi1 x g for decante origina before. the lae resuspe Tris-hf dissol‘ minute SUpern (Spect dialyz M Tris total deter; Louis isoth was a a fit mixtt 6O temperature. Serum was dispensed into an Erlenmyer flask. The gamma-globulin was precipitated by the addition of an equal volume of saturated ammonium sulfate solution dropwise while stirring with a mechanical magnetic stirrer. After all the ammonium sulfate had been added, the mixture was stirred for 20 minutes. Precipitated gamma-globulin was then centrifuged at 1,900 x g for 30 minutes at 4°C. After the supernatant was decanted, the pellet was resuspended in one-half the original volume of ammonium sulfate and centrifuged as before. This step was repeated (usually 3 times) until the last pellet was completely white. The pellet was resuspended in the smallest amount possible of 0.01 M Tris-hydrochloric acid buffer (HCl) (pH 9.0). The dissolved pellet was centrifuged at 1,100 x g for 30 minutes at 4°C. To remove residual ammonium sulfate, the supernatant was dispensed into Spectrapor membrane tubing (Spectan Medical Industries, Inc., Los Angeles, CA) and dialyzed overnight at 4°C against a large volume of 0.01 M Tris-HCl in a beaker with a magnetic stirrer. The total protein of the concentrated gamma-globulin was determined by the biuret method (Sigma Diagnostics, St. Louis, MO). Sufficient isomer I of fluorescein isothiocyanate (FITC) (Sigma Chemical Co., St. Louis, MO) was added to the gamma globulin in a 50 ml beaker to give a fluorochrome-to-protein ratio by weight of 1:100. The mixture was slowly (to prevent denaturation of the globulin) refrigeré the mixtt cm in he: (Pharmac equilibr (pH 7.3) Bee was adse Louis, 1 (pH 7.3 mg of l and all Tissue minute: time a1 um APD plasti diluti Stainj dilut: Place Napex tissr (IUte 61 globulin) stirred overnight by a mechanical stirrer in a refrigerator at 4°C. Unconjugated FITC was removed from the mixture by gel filtration through a glass column (60 cm in height x 5 cm in diameter) packed with Sephadex—25 (Pharmacia Fine Chemicals, Inc., Piscataway, NJ) equilibrated with 0.1 M phosphate buffered saline (PBS) (pH 7.3). Because of non-specific fluorescence, the conjugate was adsorbed with liver powder (Sigma Chemical Co., St. Louis, MO). The conjugate was adjusted with 0.1 M PBS (pH 7.3) to a concentration of 3.3 mg of protein/ml. Ten mg of liver powder per mg of conjugate protein were mixed and allowed to stand 1 hour with occasional stirring. Tissue powder was removed by centrifugation for 30 minutes at 20,000 x g. This process was repeated one time and the conjugate was then filtered through a 0.45 pm APD Millipore filter. Small aliquots of conjugate in plastic vials were stored at —20°C until used. Various dilutions of conjugate in 0.1 M PBS were tested in staining trials to determine the appropriate conjugate dilution for FAT. Examination by Immunofluorescence Specimens for fluorescent antibody testing were placed in embedding medium (Miles Laboratories, Inc., Naperville, IL), and quick frozen at —70°C. Frozen tissues were cut 8—10 pm thick with a freezing microtome (International Equipment Company, Needham Heights, MA) and then After fi: tissue s anti-rot section anti-rot were ap1 for 30 I minutes minute mountin and cov examine immunoi illumiI German: Site w coarse to con rotavi aS the Secti eOSin Hunt, QVale 62 and then two sections were placed on one glass slide. After fixation in acetone for 10 minutes, one of these tissue sections was stained with fluorescein-conjugated anti—rotavirus (SDSU strain) globulin and the other section was stained with fluorescein—conjugated anti—rotavirus (OSU strain) globulin. After the stains were applied, the slides were placed in a humid chamber for 30 minutes at 37°C. Then slides were washed for 5 minutes in each of 2 rinses of 0.1 M PBS and then for 1 minute in distilled water. Slides were air-dried and mounting medium pH 9 (Difco Laboratories, Detroit, MI) and coverslips were applied. Stained preparations were examined by immunofluorescence microscopy using a Zeiss immunofluorescent microscope with epifluorescent illumination and a Xenon light source (Carl Zeiss, West Germany). At least 4 sections of small intestine per site were evaluated. Specimens with intense fine to coarsely granular cytoplasmic fluorescence when compared to control tissues were considered positive (+) for rotavirus antigen. Tissues from uninfected pigs served as the controls. Light Microscopy Formalin—fixed tissues were embedded in paraffin, sectioned 6 pm thick, and stained with hematoxylin and eosin using standard histologic techniques (Thompson and Hunt, 1966). Histopathologic changes in tissues were evaluated with the use of a light microscope. In addition ocular m height ( crypts v intestir mucosal made to Additio subject scale w and a ( cuboide conside by the segmen (-) wh which villor Piece addit other Princ each M phc Chang 63 addition, small intestine was examined using a calibrated ocular micrometer to determine crypt depth and villous height (Moon et a1, 1975). Five, well—oriented villi and crypts were measured from each segment of small intestine. Because submucosal lymphoid nodules affect mucosal architecture (Mouwen et al, 1971), no attempt was made to measure villi or crypts in these areas. Additional changes in intestinal segments were subjectively graded on a negative (-) to plus three (+3) scale where (-) represented no change from the control and a (+3) represented the greatest change. Squamous, cuboidal, and low columnar epithelial cells were considered immature. Cytoplasmic vacuolation was scored by the number of vacuolated epithelial cells in each segment of intestine. Vacuolation ranged from a negative (—) which represented no vacuolation to a plus four (+4) which equaled vacuolation of approximately 100% of the villous epithelial cells. Scanning Electron Microscopy Tissues for SEM were refrigerated at 4°C until processed for SEM (usually within 24—48 hours). All additional processing was done at room temperature unless otherwise specified. Duodenum, UJ, and LI from 3 principal pigs and 1 control pig per treatment group at each time period were rinsed in each of 3 changes of 0.2 M phosphate buffer and washed for 8 hours in each of 3 changes of double—distilled water. Tissue was post—fixed in 1% 03 wash, an additior Tie (Polyscf were d0] and I-Iil thiocar treatme tissue over 3C Ti (Maser acidif' H(31/50 for 15 change dehydr ethane Vacuur Garbo] Silve- Co. 9 exami (lute acce] magnj 64 in 1% osmium tetroxide for 30 minutes. The above rinse, wash, and post-fixation steps were repeated one additional time. Tissue staining with thiocarbohydrazide (Polysciences, Inc., Warrington, PA) and osmium tetroxide were done using procedures described previously (Malick and Wilson, 1975) with the following modifications: the thiocarbohydrazide was a saturated solution; each treatment with osmium tetroxide was for 30 minutes; and tissue was washed in 3 changes of double—distilled water over 30 minutes. Tissue dehydration was done by reported methods (Maser and Trimble, 1977) except that only 2 ml of acidified dimethoxypropane (DMP) (1 drop of concentrated HC1/50.0 m1 of DMP) was applied to each tissue section for 15 minutes. The DMP was removed and each of 3 changes of 100% ethanol was applied for 15 minutes. The dehydrated ethanol treated specimens were placed in 100% ethanol in a critical point drying apparatus (Denton Vacuum, Inc., Cherry Hill, NJ) and were dried in liquid carbon dioxide. Dried specimens were attached with silver conducting paint (Acme Chemicals and Insulation Co., New Haven, CT) to aluminum SEM stubs and were examined using a Super IIIA scanning electron microscope (International Scientific Instruments, Korea) at an accelerating voltage of 15kv and at various magnifications and working distances. Images were recorded Cambridg Fee to inoce isolator Sterile fecal s. swabs w control stored ~70°C u Ea Suspenc by cent pellet at 40,' discar distil A Ritchi of dis (PTA) (BSA) depre. aPpli Tnsti 65 recorded on Polaroid Land Film Type 55 (Polaroid Corp., Cambridge, MA). Electron Microscopic Examination of Feces Fecal specimens were collected from each pig prior to inoculation and from the remaining pigs in each isolator 24, 72, and 168 hours after inoculation. Sterile plastic tubes (12 x 75 mm) were used to collect fecal samples, or if pigs were not scouring, sterile swabs were used. Cecal contents were collected from each control and infected pig at necropsy. Specimens were stored in a freezer (Revco, Inc., Deerfield, M1), at —70°C until processed. Each specimen was thawed at room temperature, suspended in Hanks' balanced salt solution, and clarified by centrifugation at 3,000 x g for 15 minutes. The pellet was discarded and the supernatant was centrifuged at 40,000 x g for 30 minutes. The supernatant was discarded and the pellet was suspended in 0.5 ml of distilled water. A negative-staining procedure as described by Ritchie and Fernelius (1968) was used. Briefly, 20 drops of distilled water, 2 drops of 42 phosphotungstic acid (PTA) pH 6.2-6.8, 1 drop of 0.1% bovine serum albumin (BSA), and 1 drop of the fecal suspension were mixed in a depression dish with a Pasteur pipette. The mixture was applied with an all—glass nebulizer (Ted Pella, Inc., Tustin, CA) to nitrocellulose coated 200 mesh grids. Specimer HU—IZA t Japan) i the PTA optimiz« Th criteri Bo analyze varianc by usir from ce level e Dr. Wi South 66 Specimens were examined immediately with an Hitachi HU—12A transmission electron microscope (Hitachi, Tokyo, Japan) at an accelerating voltage of 75kv. If needed, the PTA, BSA, or sample concentrations were adjusted to optimize spreading and contrast. The density of viral particles was estimated by criteria described by Benfield et a1 (1984). Statistical Analysis Body weights, villous heights, and crypt depths were analyzed statistically by using the one-way analysis of variance. Differences between group means were analyzed by using the Waller-Duncan K-ratio T—test. Differences from control values were considered significant at the level of p<0.05. (The statistical analysis was done by Dr. William Tucker, Agricultural Experiment Station, South Dakota State University, Brookings, SD). All with the observed strain. deveIOp . with the lengths observed strain, not have The the SD31 Pigs wi' undiges hours p Persist inocula RESULTS Clinical Signs A11 pigs in the control group and pigs inoculated with the OSU or SDSU strains survived. Diarrhea was observed in 10/14 pigs 19-36 hours P1 with the OSU strain. Four pigs inoculated with the OSU strain did not develop diarrhea until 44—48 hours PI. Pigs inoculated with the SDSU strain had incubation periods of varying lengths before the onset of diarrhea. Diarrhea was observed in 11/14 pigs 24 to 54 hours P1 with the SDSU strain. Three pigs inoculated with the SDSU strain did not have diarrhea when killed at 24 hours PI. The appearance of feces from pigs inoculated with the SDSU or OSU strains were similar. Initially, pigs with diarrhea had watery yellow feces. Curds of undigested milk in the feces were first observed 48—96 hours PI. Diarrhea with curds of undigested milk persisted until pigs were killed 168 hours PI. Pigs inoculated with the OSU strain in most instances became reluctant to drink milk shortly before the onset of diarrhea, and milk remained in the feeding bowls at the time of the next feeding. The pigs infected with the OSU strain remained anorectic until 48-96 hours PI. Vomiting 67 g was obsei the OSU : strain d' Con the expe fter ea characte Pig rotavir1 comparee pigs in pigs ha The rot gains 1 inocule by the by 144 a weig the sa pig in 152 wt inocu] of 27: body \ stati 68 was observed in 5 of 14 (36%) of the pigs infected with the OSU strain, whereas pigs inoculated with the SDSU strain did not vomit. Control pigs remained active and alert throughout the experiment and rapidly consumed their entire ration after each feeding. Control pigs had pasty golden feces characteristic of milk-fed gnotobiotic pigs. Body Weight Pigs inoculated with the OSU or SDSU strains of rotavirus had lower body weights and reduced weight gains compared to the controls (Table 1). At 168 hours PI, pigs infected with the OSU or SDSU strains and control pigs had weight gains of 16, 22, and 46%, respectively. The rotavirus-infected pigs had highly variable weight gains irrespective of the strain of rotavirus used as the inoculum. These differences in weight gain are reflected by the high standard deviations (Table 1). For example, by 144 hours PI, a pig inoculated with the OSU strain had a weight loss of 25% whereas another pig inoculated with the same virus had a weight gain of 21%. Similarly, a pig inoculated with the SDSU strain had a weight loss of 15% whereas another pig inoculated with the same pool of inoculum and kept in the same isolator had a weight gain of 27%. Because of this extreme pig to pig variation, body weights of virus infected pigs were not statistically different (p<0.05) from controls. Table 1 pigs in rotavir‘ Treatme rou Control OSU SDSU a groups 69 Table 1. Mean body weights (g i SD) for gnotobiotic pigs infected with the OSU or SDSU strains of porcine rotavirus Hours after inoculation Treatment group 0 24 48 72 96 120 144 168 Control 1254a 1295 1456 1520 1620 1652 1741 1832 i142 i142 i158 i143 i127 i153 i160 1138 OSU 1320 1323 1386 1363 1362 1386 1432 1526 i222 i255 i177 i232 i248 i265 i292 i311 SDSU 1257 1316 1309 1346 1378 1412 1464 1534 i224 i227 i220 i228 $88 $99 i112 i126 a Body weights of control, OSU, and SDSU treatment groups were not significantly different (p<0.05). He pigs he deliver useful with tl chemis1 proteir inocul. total‘ 24, 72 reduce Howeve l68 ho contrc pigs i time. before serum Chlor: cont 1:. Varied as the judged PrOceE Clinical Pathology Hematocrit values varied among pigs because some pigs had excessive umbilical cord bleeding after delivery. Therefore, hematocrit measurements were not useful as an index of hemoconcentration. Pigs inoculated with the OSU or SDSU strains had similar changes in serum chemistry values (Tables 2, 3, and 4). The total serum protein and BUN values were greater in rotavirus inoculated pigs than in controls. The elevations in total protein and BUN were greatest at 168 hours PI. At 24, 72, and 168 hours PI, serum triglycerides were reduced in virus infected pigs when compared to controls. However, results of serum triglyceride measurements at 168 hours PI were not meaningful because sera from 3 controls, 3 pigs inoculated with the OSU strain, and 2 pigs inoculated with the SDSU strain were lipemic at this time. Lipemia, which indicates insufficient fasting before the collection of serum, may have interfered with serum chemistry determinations. Sodium, potassium, chloride, cholesterol, glucose, and creatinine values for control and rotavirus infected pigs were similar. Gross Lesions The external appearance of virus—inoculated pigs varied and was independent of the strain of rotavirus used as the inoculum. Some pigs were thin and dehydrated (as judged by decreased skin pliability and prominence of bony processes) and had rough hair coats whereas others Table 2 gnotobie or SDSU __—--—— ire-e Hematoc Total p Glucose BUN Creatir Sodium. Potass Chlori Cholee Triglj ‘— l 71 Table 2. Serum chemistry and hematocrit values for gnotobiotic pigs 24 hours after inoculation with the OSU or SDSU strains of porcine rotavirus Treatment group Parameter Units Control OSU SQSU Hematocrit Z 29.0 i 1.2 29.8 i 0.2 26.4 i 0.5 Total protein gm/dl 2.5 i 0.3 2.6 i 0.1 2.6 i 0.2 Glucose mg/dl 97 i 14 110 i 5 83 i 0 BUN mg/dl 7.8 i 0.3 9.4 i 4.9 18.6 i 14. Creatinine mg/dl 1.0 i 0.1 1.0 i 0.1 1.1 i 0.1 Sodium meq/l 149 i 0.6 148 i 3.4 151 i 4.3 Potassium meq/l 5.4 i 0.2 5.8 i 0.6 5.6 i 0.7 Chloride meq/l 108 i 2.3 108 i 1.1 108 i 4.0 Cholesterol mg/dl 149 i 33 136 i 14 180 i 8 Triglyceride mg/dl 90 i 2 34 i 17 51 i 18 Data are expressed as the mean i SD. Table 3. otobio or SDSU # mains Hematocr Total p1 Glucose BUN Creatin Sodium Potassi Chlorid Cholest Triglyc N De Table 3. 72 Serum chemistry and hematocrit values for gnotobiotic pigs 72 hours after inoculation with the OSU or SDSU strains of porcine rotavirus Treatment group OS Parameter Uplpp Control SQSU Hematocrit % 27.0 i 2.3 28.3 i 0.4 25.3 i 2.0 Total protein gm/dl 2.8 i 0.3 3.0 i 0.5 2.9 i 0.2 Glucose mg/dl 116 i 26 103 i 4 110 i 17 BUN mg/dl 7.5 i 0.6 27.0 i 18.5 26.3 i 28. Creatinine mg/dl 1.1 i 0.1 0.9 i 0.2 1.1 i 0.3 Sodium meq/l 154 i 9.2 153 i 10.7 154 i 2.7 Potassium meq/l 6.1 i 0.8 6.5 i 0.7 5.7 i 0.1 Chloride meq/l 118 i 10.7 116 i 8.4 116 i 5.8 Cholesterol mg/dl 157 i 15 137 i 21 113 i 12 Triglyceride mg/dl 89 i 17 46 i 34 48 i 19 Data are expressed as the mean i SD. Table 4. gnotobic or SDSU # rams! Hematocr Total p1 Glucose BUN Creatin: Sodium Potassi Chlorid Cholest Triglyc \— Da 73 Table 4. Serum chemistry and hematocrit values for gnotobiotic pigs 168 hours after inoculation with the OSU or SDSU strains of porcine rotavirus Treatment group Parameter Upipg Control OSU SQSU Hematocrit % 32.0 i 4.6 28.4 i 2.9 27.0 i 0.0 Total protein gm/dl 2.7 i 0.3 3.3 i 0.4 3.3 i 0.0 Glucose mg/dl 151 i 8 119 i 8 105 i 24 BUN mg/dl 5.0 i 0.0 37.2 i 30.4 31.6 i 22.8 Creatinine mg/dl 1.2 i 0.1 1.0 i 0.1 1.0 i 0.0 Sodium meq/l 148 i 2.3 154 i 12.1 150 i 9.6 Potassium meq/l 5.8 i 0.0 5.2 i 0.2 5.1 i 0.3 Chloride meq/l 111 i 4.0 119 i 12.8 114 i 10.1 Cholesterol mg/dl 143 i 25 157 i 26 128 i 51 Triglyceride mg/dl 203 i 13 120 i 92 62 i 25 Data are expressed as the mean i SD. appearee fecal 31 In1 infectee The stor amounts with wa' of incor hours P pale, f PI. Th strain normal Tb Chyle i Small 5 Of the Organs Gt LYmpha Contai intest L \— V from p rOtavi PI (Tc C 74 appeared healthy except for the presence of diarrhea and fecal staining on the perineum. Internally, gross changes were similar in pigs infected with either the OSU or SDSU strain of rotavirus. The stomachs of virus-infected pigs contained various amounts of clotted milk. Ceca and colons were distended with watery yellow contents which contained small clots of incompletely digested milk in pigs killed at 72 and 168 hours PI. The small intestine in pigs with diarrhea was pale, flaccid, and thin-walled at 24, 72, and 168 hours v PI. The intestines of two pigs inoculated with the SDSU strain which did not have diarrhea at 24 hours PI were normal grossly. The majority of virus-infected pigs did not have chyle in mesenteric lymphatics 24 hours after inoculation. Small amounts of chyle were seen in the proximal one—third of the small intestine at 72 and 168 hours PI. Other organs of infected pigs were normal grossly. Gross lesions were not seen in control pigs. Lymphatics in the small intestine of control pigs contained various amounts of chyle in the proximal small intestine 24, 72, and 168 hours PI. Light and Scanning Electron Microscopic Changes Villi in each of the small intestinal sites sampled from pigs inoculated with the OSU or SDSU strains of rotavirus were shorter than control villi at 24 hours PI (Table 5). The DU was less severely affected by Table gnotol porcir H01 aft inocul 2! 16 for t lnocu Value beth 75 Table 5. Villous length (pm) in the small intestines of gnotobiotic pigs infected with the OSU or SDSU strains of porcine rotavirus Hours after Treatment Intestinal site inoculation group DU UJ LJ MI UI L1 24 Control 855 906 813 825 781 815 i4 ea i12 i6 i48 e18 osu 645 342a 501 541 594 546 i5 i44 e12 i27 ill e11 snsu 675 526a 544 537 513 422 i31 e40 e30 i58 i0 i24 , 72 Control 827 943 833 839 897 952 i7 $7 $28 e9 i63 e12 osu 614a’b 227a 124a“b 82a 194a 249a i82 e37 e19 e7 e11 i4 sosu 3543’b 311a l96a’b 155a 247a 366a 113 e23 i2 i0 i3 i7 168 Control 845 810 823 939 1016 1002 $19 i27 i27 i9 144 i64 osu 874 419a 364a 321a"b 376a 686a i42 ilO i9 e21 e10 e5 snsu 439‘?"b 322a 289a 245‘?"b 293a 640a i1 i26 i4 i9 i7 e22 Data are expressed as the mean i SE (N = 20 except for the OSU and SDSU groups at 24 and 168 hours after inoculation in which N 25). Values significantly different from control values, p<0.05. Significant difference (p<0. 05) in villous length between the OSU and SDSU inoculated pigs. villous villous differe groups 24 hou1 the OSI Villous with tl (Figure infecte controi the UJ SDSU 8' mean v SDSU s 168 ho- strain in the hours in the were 6 of pie Villi or SDE PI, b1 DU of Villi 76 villous atrophy than the other sites. The reduction in villous length in the UJ (Figure 1) was significantly different (p<0.05) when each of the rotavirus inoculated groups of pigs were compared to the control (Table 5). At 24 hours PI, mean villous lengths for pigs inoculated with the OSU or SDSU strains were not significantly different. Villous atrophy in the small intestines of pigs infected with the OSU or SDSU strains was maximal at 72 hours PI (Figure 2). At this time, villous lengths in virus infected pigs were significantly (p<0.05) shorter than controls (Table 5). Villous atrophy was most severe in the UJ, LJ, MI, and UI of pigs infected with the OSU or SDSU strains of rotavirus at 72 and 168 hours PI. The mean villous lengths in the DU of pigs infected with the SDSU strain were significantly shorter (p<0.05) at 72 and 168 hours PI when compared to pigs infected with the OSU strain or control pigs (Figures 3, 4, 5, 6, and 7). Villi in the duodenum of pigs infected with the OSU strain 168 hours PI were the same length as the control villi. Villi in the LJ of pigs infected with the OSU strain 72 hours P1 were significantly shorter (p<0.05) than villi in the LJ of pigs infected with the SDSU strain and control pigs. Villi in the small intestine of pigs infected with the OSU or SDSU strains were longer at 168 hours than at 72 hours PI, but were still significantly shorter, except for the DU of pigs infected with the OSU strain, than control villi at each site. Pigur Pig 2 Porci (arro tips 77 Figure 1. Scanning electron micrograph of the UJ of a pig 24 hours after inoculation with the OSU strain of porcine rotavirus. Notice the rounded absorptive cells (arrowhead) and the exposed lamina propria (arrow) on the tips of atrophic villi. Bar = 100 um. Figur pig ? Porci denue 78 Figure 2. Section of small intestine from the MI of a pig 72 hours after inoculation with the OSU strain of porcine rotavirus. Villi are short, fused, and focally denuded (arrow). Hematoxylin and eosin stain; bar = 10 um. ‘ Km ’ _. 16. (2m Figu Pig pore stai 79 testine from the DU of a In Section of small 3. igure F th the SDSU strain of ion w1 lat anCH 72 hours after ’ pig lined Moderately shortened villi are irus. tav porcine ro d eosin in an o Hematoxyl by columnar absorptive cells. ; bar = 100 um. stain .- 10.5 1.11. e val. can... . .2 M I I -l U I i’lto‘J’. 'u’ ,_! Figur pig 1 porci and l Hemat 80 Figure 4. Section of small intestine from the DU of a pig 168 hours after inoculation with the SDSU strain of porcine rotavirus. Villi are short and fused (arrows) and lined by low to tall columnar absorptive cells. Hematoxylin and eosin stain; bar — 100 um. Pig 1 Porci fused 100 u 81 Figure 5. Scanning electron micrograph of the DU of a pig 168 hours after inoculation with the SDSU strain of porcine rotavirus. The tips of severely shortened and fused villi have areas of exposed lamina propria. Bar = 100 um. Figu: pig 2 POrc: Ville lOO~ 82 ine from the DU of a f small intest ion 0 Sect Figure 6 lation with the OSU strain of pig 168 hours after inocu tologically normal Notice the his tavirus. porcine ro in and eosin stain; bar = Hematoxyl villous structure. 100 um. Figure 7 big 168 ‘ Porcine Control- Seen (ar 83 Figure 7. Scanning electron micrograph of the DU of a pig 168 hours after inoculation with the OSU strain of porcine rotavirus. Villous structure is similar to control villi. Epithelial cell extrusion zones can be seen (arrows). Bar = 100 um. Smal principal hours PI, infected crypts in deeper in hours PI at all si of pigs i significa crypt dep Fusi inoculate (Tables 7 intestine numerous were Obse with the with the The smal] Villous f Cont Cells in 11). The in the dj aged. C3 rOtaviruE 84 Small intestinal crypt depths in control and principal pigs were not significantly different at 24 hours PI, except for the crypt depths in the UI of pigs infected with the SDSU strain (Table 6). At 72 hours PI, crypts in pigs infected with the OSU or SDSU strains were deeper in the UJ-LI than those of controls and by 168 hours PI this difference in crypt depths was significant at all sites. Crypts at each site in the small intestine of pigs infected with the SDSU strain 168 hours PI were significantly deeper, except in the L1, when compared to crypt depths in pigs infected with the OSU strain. Fusion of villi in the small intestine of rotavirus inoculated pigs was observed 24, 72, and 168 hours PI (Tables 7, 8, and 9). All of the segments of small intestine were affected, but fused villi were most numerous in the UJ-UI (Figures 8, 9, and 10). Fused villi were observed more frequently in the DU of pigs inoculated with the SDSU strain (9 of 9 pigs) than in pigs inoculated with the OSU strain (2 of 9 pigs) at 72 and 168 hours P1. The small intestine of control pigs had a few areas of Villous fusion 24 and 72 hours PI. Control pigs had vacuolated absorptive epithelial cells in the UJ—LI and less commonly in the DU (Figure 11). The amount of epithelial cell vacuolation increased in the distal small intestine and decreased as the pigs aged. Cytoplasmic vacuolation in the small intestine of rotavirus infected pigs, particularly at 168 hours PI, was Table 6. gnotobioti porcine rc _________ Hours after inoculatie 24 Dat the OSU in Which V P<0.0S. bELWEQHK Table 6. 85 Crypt depths (pm) in the small intestines of gnotobiotic pigs infected with the OSU or SDSU strains of porcine rotavirus Hours Treat- after ment Intestinal site inoculation group DU UJ LJ MI UI L1 24 Control 97 98 9o 77 91 85 $6.8 $0.8 $2.6 $11.6 $0.5 $0.9 osu 94 89 92 75 74 8o $3.4 $1.4 $6.2 $0.6 $10.0 $0.5 snsu 85 82 71 70 66a 70 $1.6 $1.1 $1.4 $0.5 $2.4 $1.8 72 Control 120 105 93 93 89 101 $1.5 $1 7 $0.0 $0.1 $3.9 $0.6 030 122 122 135 143a 1288 121 $4.4 $5 1 $5.6 $1.5 $2.1 $3.0 snsu 125 130 121 118 124a 109 $0.0 $2 1 $0.6 $9.4 $3.7 $6.0 168 Control 113 107 101 102 89 98 $4.7 $6.9 $4.2 $1.4 $2.1 $8.3 osu 1518’b 165"?"b 149"?"b 153a"b 139a;b 136a $2.9 $2.0 $4.3 $9.8 $4.3 $3.7 sosu ISIa’b 1898 b 1935"b 179a"b 1605"b 14oa $4.3 $2.4 $6.9 $0.8 $3.1 $1.1 Data are expressed as the means i SE (N = the OSU and SDSU groups 24 and 168 hours after inoculation in which N = 25). 20 except for a Values significantly different from control values p<0.05. b Significant difference (p<0.05) in crypt depth between the OSU and SDSU inoculated pigs. £11-43. .1 c, -. A DWQW HO DWO ®£u SUHB COHUQHSUCCH hmuwfi mDHH>muCH QCflUHOQ HO Cfimhum fl mefiUCHM UHQOUWOHUflE U£WHQ .m QHQQH mHDOfi {N meQ OHHOHQOUOCw WC OCHumGUCfi HHQEw MSU C 86 .memfio ummummuw msu wcflucwmoumwp m m tam Howucoo Scum mwcmfio 0c 0 Any “m .N .H .1 mo mflmwn onu co meoom mums mufiusuweaw cam .COHme .u:oE£oMuoo n .me Loom Eowm muHDmmu wwumummmm mEEoo m umHHmo Hawaofiuflmm mDOHHH> wsu we NOOH mamumfiwxoumam mo Cowumaosom> 0 Any ou cowumaosum> o: 0 any scum wcfimcwh .COHuwHosow> mo mwummw mSu co wwhoom mmB cowuwaosom> ofiEmmamoumo m l ml ml mm mm l ml ml mm ml—H l ml ml ml mN m mfiN mq mH ml_H Hulk. H ml ml mm ml H mlfl ml mm ml l ml ml ml ml m mm mq ml mH Hp H ml ml mm ml H mH ml mm ml 1—.. ml ml ml—H ml H mN mm ml mH H: l ml ml mm ml l mH ml mm ml l ml ml mH ml H mr—H mN ml mA_H HIWH. m ”l ”l Hm ”I 1H ml ml mm mH H ml ml mH ml H mH mH ml ml h}: l l l H l l ml ml ml mH l ml mI mI ml l ml ml ml ml DD swam l ”l ”l Hm mm l ml ml mm mm l ml ml mH mm q m4N mq mA—H ml HWH l ml ml N mm l ml mI mN mm l ml ml mH mm q mm mq ml ml HD l l l mH mm l mH mH mN mm l ml ml mH mm q mm mq mI—n. ml HHA l mu” ml mm mm l mH ml mm mm l ml ml mH mm m mm mH ml ml H411” N ml ml mN mm l mN ml mm mm m ml ml mm mm H mN mN ml ml HID l mN ml mN mm l ml ml ml m|_H l mN ml mH mN N ml ml ml ml DO Dmo l ”l ”l ”l l ml ml ml—H l ml ml ml q mq mq mq H1.” l ml ml ml l mH mH mH l ml ml ml q mm mm m¢ HD I ml ml l l mufl mH ml l ml ml ml m mN mN mm H: l ml ml ml l mH mH ml l ml ml ml m mH mH mN HJWH. l ml ml ”l l ml ml ml l ml ml ml N mH mH mH HJD l l l l l ml ml ml l ml ml ml A—H ml ml ml :9 HOIHUEOO Eflwamfiuflmm mDOHHH> commsw unwesomuwv coauwaodom> ouflm msoyw anSumEEH AmsoHHfi> nHHwo m>HumHomfl< maaoo m>HumHOmfi< HmcflumoucH ucoEummHH mDMH>muou ocfiosom mo chHum swam Ho Dmo map SuHB coaumadoocfi Mmumm muses «N wwwa ofiuoanouocw mo mcflumwucfl HHmEm can CH mwcwpcww osmoowoyofla unwed .m waan mDHfl>muOH QCHUHOQ MO mCHQHum DQO HO DmO mnu SUHB COwumHDUOEH Mmuww mhfiOfi Nh meQ UHUOHQOUOCw MO GCHquUCH HHmEm OSU CH meHECHM UHQOUmOHUflE u£wwd .m Omng 87 .mwcmno ummumwhw mfiu wcflucwmmhawu m m Ucm HOHucoo Eouw mwcmno 0G u Auv um .N .H .n mo mammn wfiu co wmhoom OHwB hufludumEEfl wcw .cOflmsw .ucmfinomumo 3 .mfla Lomw Eoum muasmwh mmumumaom mEEoo m “mHHwo HmHHwfiuHmm mDOHHfl> mnu we NOOH hawuwfiwxoyamm mo coaumacsom> u Aqv ou Coaumaosom> 05 n AIV Eouw wfiwwcmw .aowumaosum> mo mwhwmv «Lu so wmpoom mmB COHuMHodom> UHEmmHmoumo m a INMMN v—INMMI—Iv—I o. n—I I HA HD .. o. a a a a a - H | u—I I N N r—II—ImNt—IN I I o. lI—I INMMI—IN mmmmmm I Iv—I I I Hv—Ir—Ir—Ir—I Nv—Ir—Imm r—IMNNx‘I'fl' I ll—Ir—II—I H v- A v—I I I v—Ir—INMM o. .- r—T : O DQO n o. c. o. o. a a u. n a o. o. a. n p. a a a INNM Iv—Iu—I Iv—INNNr—I I l I I r—IMMMMN I—II—INNI—Iv—I IMMMI—I IMMMMN Ir-Ir—IMr—I lr-Imm Ir-IN a I c. a - I o. - Dmo I I I I I .. p. a. n o. lr—INNM NNNr—II—IN D Q IHMMQ‘Q’ n a IHNM soflmsm usmfifiomuwv Coaumflodow> muam adouw nmhsumEEH AmDOHHH> AHHwo m>HuQH0mn< «Hawo m>fluaHOmn< HwCHuwmucH quEumwa mSHH>mu0H onogom mo mchuum Dmom Ho Dmo msu suHB cowumHDQOCH Mouwm muse: Nu mmflm ofluOflnouocm mo mcfiummucfl HHmEm mfiu CH mwzfiwcfiw owaoomouoflfi ucwfld .w wanwe mDHH>quH @CHUHOQ MO mCHwHum Dmc mHDOS woa mwfim UHUOHQOuocw wo OCHumquH HHmEm ®£u CH m m HO DmO ®£u fiuHE COflumHDUOCH Mmuwm WGHQCHH UHQOUWOHUHE unwflq .m OHDQH 88 Anv EOHM mcfiwcmh .COHumHosom> mo mmummw mfiu co wmuoow mm: coaumaodum> oHEmmHaouzo .QMGMSU ummumwww mfiu wcfiucmmmuamh m m cam HouucOu Eoyw wwcmfio on u Alv “m .N .H .u mo mammn wan no Umpoum meB huflHSumEEH cam .EOHmSm .uCQEfiumuwQ n .me some Eowm muaswmu mmumumaom mEEou m mmHHoo HmHHmnuHam mDOHHH> wfiu mo NOOH mamumfiflxOHmmm mo COHuMH050m> u Aqv ou Soauwaosom> on n m .. I H l o. ., a a v. a a a a Il—Ir—I I I I .. INMMr—Ir—I r—Iv—I NI MI I I NMMMMr—I NMNMN o. o. o IN mNNF-IF'I a .- Nle—INr—I NNMNNI—I c. p. o. I I | | l IMNC’Ir—II—I INMNN INr—I nan-\— lNCJNNr—I r-INMMMr—I a o. n p. p. n. u. n Hg - l - H: I o..- I .o.. I A‘. I .c... I r—I “a-.. a a o. I I v—Ir—I I H I Ir-Iy—I u—Ir—Ir—Ir-Ir-I r—I I I I I "J D n n o. I r—I lu—Iu—Ir-I H D a a u Ir—Ir—I I I h :3 I I I I l I—Ir—Il—II—Ir—IN I I I I IHCVWM¢ HF4FI Ir4OJA [chawr H 2 I—Ir-Ir—INMM Dmom Dmo HowucOo ESMHmSmew mDOHHH> COHmDm 0H5 NE a u H pmsoHHH> n unmafiomuow Goaumaosum> muam adopw Hawo 0>HuQHOmQ< «Hamo w>HumHomn< HmcfiuwmucH quEuwwHH msuw>wu0M wcfiouoa mo mcflmuum swam Ho Dmo wfiu nufl3 Goaumasoocfl wmuwm muses wcH wwwa oauownouocw mo wcfiumquw HHmEm wfiu CH mwcfl©CHm oHaoomouoHE uLwHA .m manme Figure from th SDSU st Stain; 89 Figure 8. Villous fusion in a section of small intestine from the LJ of a pig 72 hours after inoculation with the SDSU strain of porcine rotavirus. Hematoxylin and eosin stain; bar = 10 um. Figure intesti inocula Loss of of dema exPosed (arrow) 9O ‘9‘» ‘ u. ~ o’.;:‘; r I. I 5... 3‘ "j w ' v¢_ . ‘I I . t b (4’ 1‘ r' 1' L r F' v Figure 9. Scanning electron micrograph of small intestine from the LJ of a pig 168 hours after inoculation with the SDSU strain of porcine rotavirus. Loss of absorptive cells creates a sharp line of demarcation (arrowheads) bordering an area of xposed lamina propria. An area of villous fusion (arrow) can be seen. Bar = 10 um. Figure 1 168 houx POrCine 100 um. 91 Figure 10. Villous fusion in a section of UJ from a pig 168 hours after inoculation with the OSU strain of porcine rotavirus. Hematoxylin and eosin stain; bar — 100 um. Figure 1 the LI ( media a] bar = 1( 92 rm an": :9 Q” vi 1..l.4¢o swan) Mossy o!&aau'm_ v Us: Most of the absorptive cells lining villi in Figure 11. the LI of a control pig 24 hours after inoculation with Hematoxylin and eosin stain; media are vacuolated (+4). = 10 um. bar much les 7, 8, at Ror cells or observed pigs at much pig detachme with exp numerous SDSU str adjacent disorgan: microvili microvilj detachmex With the hours P1 With the at 72 and inoculate cell deta uncommon . time. C01 or exposec Immat 93 much less conspicuous (Figure 12) than in controls (Tables 7, 8, and 9). Rounding and detachment of absorptive epithelial cells on the tips and sometimes the sides of villi were observed in the small intestine of rotavirus infected pigs at 24 hours PI (Table 7). At this time, there was much pig-to—pig variation in the amount of cell ’ detachment. Rounding and detachment of absorptive cells with exposure of the underlying lamina propria were most numerous in the UJ—UI of pigs infected with the OSU or SDSU strains 72 hours PI (Table 8). Microvilli in areas adjacent to rounded or detached absorptive cells were disorganized or reduced in number. Areas devoid of microvilli were often sharply demarcated from normal microvilli on the same absorptive cell. Absorptive cell detachment was observed in the duodenum of pigs infected with the OSU strain 24 hours PI, but not at 72 or 168 hours PI (Tables 7, 8, and 9). In contrast, pigs infected with the SDSU strain had cell detachment in the duodenum at 72 and 168 hours PI, but not at 24 hours. Pigs inoculated with the OSU strain did not have epithelial cell detachment 168 hours PI, and cell detachment was uncommon in pigs infected with the SDSU strain at this time. Control pigs did not have areas of cell detachment or exposed lamina propria (Figures l3, l4, and 15). Immature villous epithelium lining the small intestinal villi of rotavirus infected pigs was observed Figure cells after rotavj 94 -- x‘ i . I ' “$6. ‘1'! ‘5’?! Figure 12. Immature cuboidal to low columnar absorptive cells line an atrophic villus in the UJ of a pig 24 hours after inoculation with the OSU strain of porcine rotavirus. Hematoxylin and eosin stain; bar = 10 um. Figure POSt-iI bar = j 95 Figure 13. Duodenum of a control pig killed at post—inoculation hour 24. Hematoxylin and eosin stain; bar = 100 um. 96 A L Figure 14. Scanning electron micrograph of the duodenum of a control pig at post—inoculation hour 24. Long slender villi have prominent transverse fissures (arrows). Bar = 100 um. 97 Figure 15. Scanning electron micrograph of the lower ileum of a control pig at post—inoculation hour 24. Villi are broader in the lower ileum when compared to villi in the duodenum (Figure 14). Enterocytes bulge into the intestinal lumen giving a cobblestone appearance to the mucosal surface. Bar = 100 um. at 24 a1 majoritj epithel: As: intestil (Figure transve1 of cont] (Figure changes lung, 1: tonsil, infectec of rote in ente small j €ntero< MI, LJ, iDOCUlg was det 72 hon} Strain, the DU hOUIS, 98 at 24 and 72 hours PI (Figure 12). By 168 hours, the majority of Villi were lined by low to tall columnar epithelial cells (Tables 7, 8, and 9). Aside from the occasional fused villi, small intestine of control pigs was histologically normal (Figure 13). Villi were long and slender with prominent transverse fissures (Figure 14). Villi in the lower ileum of controls were broader and had prominent enterocytes (Figure 15) and numerous goblet cells. Histopathologic changes were not observed in the cecum, colon, rectum, lung, liver, heart, pancreas, spleen, brain, thymus, tonsil, and mesenteric lymph node of control or rotavirus infected pigs. Immunofluorescence Pigs inoculated with either the OSU or SDSU strains of rotavirus had finely granular cytoplasmic fluorescence in enterocytes lining the sides and tips of villi in the small intestine (Tables 10 and 11). Immunofluorescent enterocytes were most consistently detected in the UJ, MI, LJ, UI, and LI of pigs 24 and 72 hours after inoculation with either rotavirus strain. Viral antigen was detected in the DU of pigs 24 hours (5 of 5 pigs) and 72 hours PI (2 of 4 pigs) after inoculation with the OSU strain. Immunofluorescent cells were seldom observed in the DU of pigs 24 hours (1 of 5 pigs) or 72 hours (0 of 4 pigs) after inoculation with the SDSU strain. By 168 hours, immunofluorescence was rarely observed in the Table l( gnotobic porcine Intestir site DU UJ LJ MI UI LI * . specific separate 99 Table 10. Immunofluorescence in the small intestine gnotobiotic piglets infected with the SDSU strain of porcine rotavirus of Intestinal Hours after inoculation site 24 12 DU -, -, -, +, -* -, -, -, - -, —, -, -, UJ -, +, +, +, — +, —, +, — —, —, -, —, LJ -, +, +, +, - +, +, +, + -, —, -, —, MI -, —, +, +, - +, +, +, + —, —, -, -, UI +, —, -, +, - +, +, +, — -, -, -, -, LI +, —, +, +, - +, —, -, - -, -, -, —, * - = No specific fluorescence for rotavirus; + = specific immunofluorescence for rotavirus; a comma separates results obtained from each pig. Table l gnotobi porcine Intesti site DU UJ LJ Ml Ul LI * specif separa 100 Table 11. Immunofluorescence in the small intestine of gnotobiotic piglets infected with the OSU strain of porcine rotavirus Intestinal Hours after inoculation site __ 72 DU +, +, +, +, +* -, -, +, + -, -, -, -, - UJ +, -, -, +, + +, +, +, + -, -, -, —, - LJ +, +, -, +, + +, +, +, + -, -, -, —, — MI +, —, +, +, + +, -, +, + -, —, -, -, — UI +, -, +, +, + +, —, +, + -, -, —, -, — L: +, -, +, +, + +, —, +, + -, —, +, —, — * - = No specific fluorescence for rotavirus; + = specific immunofluorescence for rotavirus; a comma separates results from each pig. small i strain. A fluores hours a additic contail fluore: inocul immuno of one S frozen lymph from c immunc antig« 101 small intestine of pigs infected with either virus strain. A few surface enterocytes with cytoplasmic fluorescence were observed in the cecum from one pig 24 hours after inoculation with the OSU strain. Two additional sections of cecum from this pig did not contain immunofluorescent cells. Specific immuno- fluorescence was not seen in the cecal mucosa from pigs inoculated with the SDSU strain, but a few detached immunofluorescent cells were observed in the cecal lumen of one pig 24 hours after inoculation. Specific immunofluorescence was not observed in frozen sections of stomach, tonsil, spleen, mesenteric lymph node, colon, or rectum. Frozen tissue sections from control pigs did not have specific immunofluorescence when stained for porcine rotaviral antigen. Electron Microscopic Examination of Feces Feces collected from control and principal pigs before inoculation did not contain viral particles. At 24 hours PI, most virus—inoculated pigs had +2 to +3 rotavirus particles in their feces (Table 12). Two pigs, one inoculated with the OSU strain and another with the SDSU, did not have clinical signs of diarrhea at 24 hours PI, but rotavirus particles were identified in cecal contents collected at necropsy. Cecum and colon in these pigs were dilated with fluid yellow contents indicating Table 12 examinat infectec M Neg +1 +2 +3 +4 Total n of Spec ‘- a obtaine b C Was est Squarej 102 Table 12. Results of direct electron microscopic examination of feces or cecal content from pigs infected with the SDSU or OSU strain of porcine rotavirus . a Number of spec1mens 24 hrsb M 168 hrs EM score we M 9_s_U M gs_U fig Neg 6 6 0 l 2 1 +1 0 l l l 0 2 +2 3 4 l 3 3 2 +3 5 3 7 4 0 0 1 +4 9 9 2 2 .0. 9 Total no. of specimens l4 l4 9 9 5 5 a Viral particles were not observed in feces obtained before rotavirus inoculation. b Hours after inoculation. C The number of rotavirus particles per grid square. was estimated and rated as +1 (0 to 5 virus particles/grid square), +2 (6 to 20), +3 (21 to 100), or +4 (>100). that d allowe 24 hou remain V virus feces strait 72 box fluid infect to de< seen : PI. 1 hours compl. parti colle inocu were 103 that diarrhea would have occurred had these pigs been allowed to survive. Virus particles were not detected at 24 hours after inoculation in the feces from the remainder of the non—diarrheic pigs. Virus particles had been seen in the feces of all virus infected pigs by 72 hours after inoculation. The feces of one diarrheic pig inoculated with the SDSU strain contained virus particles at 24 hours, but not at 72 hours PI. The feces from this pig were becoming less fluid so it may have been recovering from the rotavirus infection. The number of rotavirus particles in the feces began to decrease by 168 hours PI. Virus particles were not seen in the feces of 3 pigs (2 OSU, l SDSU) at 168 hours PI. Feces or cecal content collected at 24, 72, or 168 hours PI contained approximately equal numbers of complete and incomplete virus particles. Rotaviral particles were not observed in the fecal samples collected from control pigs 24, 72, or 168 hours after inoculation. In general, the virus excretion patterns were similar for both rotavirus strains. F gnotob strain signs caused duoden with t freque strain V strain findin McAdar after rEport colost McNult Graham report Narita r0tavi- (1980) DISCUSSION Findings in this study indicate that in 3-day-old gnotobiotic pigs serotype 1 (OSU) and serotype 2 (SDSU) strains of porcine rotavirus cause different clinical signs and histopathologic lesions. The SDSU strain caused more severe villous atrophy and fusion at the duodenal—jejunal flexure when compared to pigs inoculated with the OSU strain. Pigs infected with the OSU strain frequently vomited whereas pigs infected with the SDSU strain did not vomit. Vomiting by gnotobiotic pigs infected with the OSU strain but not the SDSU strain is consistent with the findings of other investigators (Theil et al, 1978; McAdaragh et al, 1980; Graham et al, 1984). Vomiting after infection by some strains of rotavirus has been reported in conventional (Bohl et al, 1978), colostrum—deprived (Lecce et al, 1976; Pearson and McNulty, 1977), and gnotobiotic pigs (Theil et al, 1978; Graham et al, 1984). In other instances, no vomiting was reported (Tzipori et al, 1978; McAdaragh et a1, 1980; Narita et a1, 1982). In fact, a strain of porcine rotavirus (B—3l7) studied by Torres—Medina and Underdahl (1980), has never been observed by these investigators to 104 cause v in this occurre differe: infecti« caused l occurre< 2 than 5 More rec people, vomiting This is al (l984 Signs ca The OSU Of human all othe Strain o 31, 1984 Used in . VOm: gaStroin1 chemorem Present 5 severe ir the SDSU FuthErnor 105 cause vomiting in gnotobiotic pigs. Therefore, findings in this and other studies suggest that differences in the occurrence of vomiting are related to rotavirus strain differences. Epidemiologic studies of human rotavirus infections also indicate differences in clinical signs caused by different strains of rotavirus. Vomiting occurred more frequently following infection by serotype 2 than serotype 1 human rotavirus (Yolken et al, l978b). More recently, Uhnoo and Svenson (1986), reported that in people, subgroup II rotavirus strains caused diarrhea and vomiting of greater severity when compared to subgroup I. This is in contrast, however, to the findings of White et a1 (1984), who could find no differences in clinical signs caused by different subgroups of human rotavirus. The OSU strain of porcine rotavirus and the DS—l strain of human rotavirus are classified as subgroup I, whereas all other human rotavirus strains and the Gottfried strain of porcine rotavirus are subgroup II (Hoshino et al, 1984). The subgroup specificity of the SDSU strain used in the present study has not yet been identified. Vomiting may be caused by irritation of the upper gastrointestinal tract or by stimulation of chemoreceptors in the medulla (Ganong, 1983). In the present study, mucosal histopathologic lesions were most severe in the upper small intestine of pigs infected with the SDSU strain, yet none of these pigs vomited. Futhermore, rotaviral antigen was not detected by IF in the stoma strain no in other 1977; The associate mechanism tract. In t the effec course of strains w temperatu were rand Pig-tO-pi infection underwent inoculati Prevent a Occur aft Uude infected neCIOPSiE SDSU virL other in\ 1980). I InfectioT l978)’ C( 106 the stomachs of pigs infected with either rotaviral strain nor has it been detected in the stomachs of pigs in other studies (McNulty et al, 1976a; Davidson et al, 1977; Theil et al, 1978). This suggests that vomiting associated with rotavirus infection may be initiated by mechanisms other than irritation of the gastrointestinal tract. In the present study, attempts were made to minimize the effects of environmental and host factors on the course of the rotaviral infections. The OSU and SDSU strains were studied while keeping diet, environmental temperature, and housing constant. Experimental animals were randomly assigned to treatment groups to account for pig-to-pig variation in susceptiblity to rotavirus infection. Furthermore, the OSU strain of rotavirus underwent four gnotobiotic pig passages prior to inoculation of experimental animals. This was done to prevent attenuation, i.e. loss of virulence which may occur after passage in cell culture (Bohl et al, 1984). Under these experimental conditions, all of the pigs infected with the OSU or SDSU strains survived until necropsied. This lack of mortality caused by the OSU and SDSU virus strains is in agreement with the findings of other investigators (Theil et al, 1978; McAdaragh et al, 1980). Mortality associated with porcine rotavirus infection has been reported in conventional (Bohl et al, 1978), colostrum—deprived (Lecce et al, 1976; McNulty et al, 19] Narita experit host 01 differe enviror infect: rotaviJ Woode, Newsome rotavi: colost to l- (1; used h (persm When 31 Virulm factor D are ev hetero 1986). of rot (bOVin HOWeve rOtavi into a 107 a1, 1976), and gnotobiotic pigs (Tzipori et al, 1978; Narita et al, 1982). However, in these studies experimental conditions differed so it is possible that host or environmental factors and not rotavirus strain differences influenced the observed mortality. Age, environmental temperature, and intercurrent bacterial infection have been shown to influence the severity of rotavirus infections (Woode et al, 1976b; Crouch and Woode, 1978; Tzipori et al, 1980b; Torres-Medina, 1984; Newsome and Coney, 1985). When a strain of porcine rotavirus which caused high mortality in l-day—old colostrum-deprived conventionally-derived pigs was given to l-day—old gnotobiotic pigs kept under the conditions used in this experiment, no mortality was observed (personal data). Therefore, caution must be exercised when suggesting that field strains of rotavirus differ in virulence, particularly because environmental and host factors may influence the severity of the disease. Differences in virulence between rotavirus strains are evident when rotaviruses are inoculated into a heterologous host (Bridger and Brown, 1984; Offit et al, 1986). For instance, the SA—ll strain (primate origin) of rotavirus was more virulent than the NCDV strain (bovine origin) when inoculated orally into newborn mice. However, evidence indicating that field strains of rotavirus are more virulent than others when inoculated into a homologous host such as in the present study is scant (Cast1 (Bridg Carpic indica chara< isolat differ ligate The a1 of vi] virule it may equate villor calves EXperi intrav Chlora shown Chlora have i I the 08 than t Theil and Va 108 scant (Woode, 1979) and often based on clinical reports (Castrucci et a1, 1983) or personal communications (Bridger, J. C. in Reynolds et al, 1985). A report by Carpio et al (1981b) is frequently cited as evidence indicating that field isolates with different virulence characteristics exist. In their report, four field isolates of bovine rotavirus consistently caused different amounts of villous atrophy when inoculated into ligated intestinal loops in colostrum-deprived calves. The authors concluded that the difference in the severity of villous atrophy corresponded to differences in virulence among the rotavirus isolates tested. However, it may be incorrect to assume that villous atrophy is equated with virulence. Reynolds et a1 (1985) observed villous atrophy in clinically normal (non-diarrheic) calves infected with rotavirus. The calves used in the experiment by Carpio et al (1981b) were maintained on intravenous chloramphenicol throughout the experiment. Chloramphenicol administered to healthy calves has been shown to induce diarrhea (Huffman et al, 1981). Chloramphenicol or the rotavirus strains, or both, may have influenced the amount of villous atrophy observed. The incubation periods after inoculating pigs with the OSU or SDSU strains, were longer and more variable than those previously reported (Crouch and Wood, 1978; Theil et al, 1978; McAdaragh et al, 1980). The prolonged and variable incubation periods were probably due to the lower c Previor diarrhe suspene 1978; l rotavim incubat do not et al, Va respon: atrophj OSU an. infect but 10: infect signif strain hours becaus differ times. R When C Variab and in Variat 109 lower dosage of rotavirus used in the present study. Previous investigations on the pathogenesis of rotaviral diarrhea caused by the OSU or SDSU strains used 102 suspensions of intestinal or fecal filtrate (Theil et al, 1978; McAdaragh et al, 1980). Decreasing dosages of rotavirus have been shown to increase the length of the incubation period before the onset of clinical signs but do not affect the severity of the clinical disease (Woode et al, 1976b). Variation in incubation periods may have been responsible for the differences in the amount of villous atrophy observed in the UJ—UI of pigs infected with the OSU and SDSU strains. Villi in the UJ-UI of pigs infected with the OSU strain were shorter at 72 hours PI but longer at 168 hours PI when compared to those of pigs infected with the SDSU strain. Glandular crypts were significantly deeper in pigs infected with the SDSU strain than in pigs infected with the OSU strain 168 hours PI. Pigs infected with the OSU or SDSU strain, because of different incubation periods, may have been at different stages of the infection when studied at these times. Rotavirus—infected pigs had reduced body weights when compared to controls. Weight gains were highly variable even when pigs were kept in the same isolator and inoculated with the same pool of virus. Pig—to—pig variation in body weight gain also was reported by Woode (1979) R but ma instan intest rotavi protei (Offit qualit enzyme of the 1976; affect recent primat 2.0, l inact: 1985) intes' entert rotav: infec 1972) Susse With the 1 110 (1979). Reasons for the pig—to-pig variation are not known, but may be related to host physiological factors. For instance, the availability of trypsin in the small intestine might affect the expression of virulence by rotaviruses. It is now clear that a trypsin-sensitive protein plays a major role in gastrointestinal virulence (Offit et al, 1986). It has been suggested that qualitative or quantitative differences in intestinal enzymes (trypsin) may be responsible for the resistance of the human neonate to rotaviral diarrhea (Holmes et al, 1976; McClean and Holmes, 1981). Gastric pH might also affect the clinical expression of rotavirus infection. A recent report indicated that three bovine and several primate-origin rotaviruses are rapidly inactivated at pH 2.0, but inactivated at a much slower rate at pH 3.0; inactivation at pH 4.0 was minimal (Weiss and Clark, 1985). The effect of host factors such as gastric pH, intestinal trypsin content, and the availability of enterocyte receptors on the expression of virulence by rotaviruses deserves further study. Serum biochemical changes that occur in rotavirus infected pigs seldom have been reported (Mouwen et al, 1972). In the present study, elevations in BUN which suggested dehydration were detected in pigs inoculated with the OSU or SDSU strains. However, as evidenced by the large standard deviation of the mean, elevations in BUN wer pathoge has bee protei: Serum 1 pigs wl presen' condit to nor trigly of the E in the regart This ; total is is more in ch (Tall Straf in t1 ShOr' isol give freq lll BUN were detected inconsistently. Another enteric pathogen of pigs, transmissible gastroenteritis virus, has been reported to cause elevations in BUN and total protein (Cornelius et a1, 1968; Drolet et al, 1984). Serum triglycerides were decreased in rotavirus infected pigs when compared to controls. Higher levels of fat are present in the feces of piglets with "white scours", a condition presumably caused by rotavirus, when compared to normal pigs (Mouwen et al, 1972). Decreased serum triglycerides may be related to the malabsorptive state of the damaged small intestine. Serum electrolyte values of rotavirus infected pigs in the present study did not differ from the controls regardless of the strain of rotavirus used as inoculum. This finding, coupled with the elevations in BUN and total protein suggests that, when dehydration occurs, it is isotonic. Isotonic dehydration also has been reported more frequently than hypertonic or hypotonic dehydration in children hospitalized with rotavirus infection (Tallette et al, 1977; Rodriguez et a1, 1977). An important finding in this study was that the SDSU strain of rotavirus produced more severe villous atrophy in the upper small intestine. Villi were significantly shorter in the DU of pigs inoculated with the SDSU isolate at 72 and 168 hours PI when compared to pigs given the OSU strain. Fusion of villi also was more frequently seen in the duodenum of pigs infected with the SDSU st is the mammali histope differe P: villou instan fusion wherea villou McAdaI used V strait B-3l7 and f EXpla the f Pigs hours were Stra: SDSU dete the inte 112 SDSU strain than pigs infected with the OSU strain. This is the first time that different serotypes of the same mammalian rotavirus have been shown to cause histopathologic lesions of different severity in different sites in the small intestine. Previous investigators disagreed about the extent of villous atrophy in the upper small intestine. For instance, McAdaragh et a1 (1980) observed atrophy and fusion of villi in the duodenum as early as 24 hours PI whereas Torres-Medina and Underdahl (1980) found no villous atrophy in the duodenum. In the experiment by McAdaragh et a1 (1980), the SDSU strain of rotavirus was used whereas Torres-Medina and Underdahl used a field strain (serotype unknown) of porcine rotavirus designated B-3l7. Differences in the severity of villous atrophy and fusion in the upper small intestine might be explained by rotavirus strain differences as suggested by the findings in the present study. Viral antigen was consistently detected in the DU of pigs infected with the OSU but not SDSU strain 24 and 72 hours PI. This was unexpected since histologic lesions were most severe in the DU of pigs infected with the SDSU strain. Three of the pigs killed 24 hours PI with the SDSU strain did not have diarrhea. Viral antigen was not detected in the small intestine from 1 of these pigs and the other two only had immunofluorescence in a few intestinal segments. It is possible that duodenal entero: became could' M differ rotavi predil pathog 132 (r intest ] McNul' some clini The G isola sugge Mixec rota\ Rodrf infec may . eith 8988 Pore atrc Snal 113 enterocytes in the pigs infected with the SDSU strain became infected between 24 and 72 hours PI, sloughed, and could no longer be detected at 72 hours PI. McNulty et al (1983) were the first to report different intestinal tropisms for different strains of rotavirus. Avian rotavirus strain ch 1 (group A) had a predilection for the duodenal mucosa of specific pathogen—free chickens, whereas avian rotavirus strain ch 132 (non—group A) replicated best in the mid-small intestine. Based on the differences in intestinal tropism, McNulty et a1 (1983) suggested that mixed infections by some strains of avian rotavirus may cause more severe clinical disease than infection by either virus alone. The Gottfried strain of porcine rotavirus was originally isolated from a mixture of rotaviruses (Bohl et al, 1984) suggesting that mixed infections occur naturally in pigs. Mixed infections by different electropherotypes of human rotavirus also have been reported (Spencer et a1, 1983; Rodriguez et al, 1983). It is possible that mixed infections in pigs by two or more strains of rotavirus may cause more severe clinical disease than infection by either strain alone. The findings in the present study suggest that mixed infections by serotypes l and 2 of porcine rotavirus might result in more severe villous atrophy with the serotype 2 rotavirus producing the upper small intestinal lesion. This hypothesis deserves to be tested H findin (Pears Woode, Underd report first rotavi' McAdar. villou contra after : porcine In the PI, so al (19? Gr lesions than tl lamina Pigs We reporte In and cec and has 114 tested. Histopathologic and scanning electron microscopic findings were similar to those reported previously (Pearson and McNulty, 1977; Theil et al, 1978; Crouch and Woode, 1978; McAdaragh et al, 1980; Torres-Medina and Underdahl, 1980; Narita et al, 1982). However, previous reports disagree about the time at which villous fusion first appears after inoculating gnotobiotic pigs with rotavirus. Theil et a1 (1978), using the OSU strain, and McAdaragh et al (1980), using the SDSU strain, observed villous fusion in gnotobiotic pigs by 24-48 hours PI. In contrast, villous fusion was first observed 4.5 days after inoculating pigs with a field strain (B-317) of porcine rotavirus (Torres-Medina and Underdahl, 1980). In the present study, villous fusion was seen at 24 hours PI, so results are in agreement with findings by Theil et a1 (1978) and McAdaragh et al (1980). Gnotobiotic pigs in the present study did not have lesions or specific immunofluorescence in organs other than the small intestine. Immunofluorescence in the lamina propria and mesenteric lymph node of gnotobiotic pigs was not observed in the present study, but has been reported by others (Theil et al, 1978). Immunofluorescence was observed in the cecal mucosa and cecal lumen of one pig infected with the OSU strain and one pig infected with the SDSU strain at 24 hours PI and has been observed in the cecum of gnotobiotic pigs by other i 1982). immunof intesti and MCI 1980). Tl the OSI simila al, 19 Torres of rot were s of rot could A (Torre detect a long fluOn infec cells (Thei fluor Peric Strai 115 other investigators (McAdaragh et a1, 1980; Narita et al, 1982). In general, the absence of histologic lesions and immunofluorescence in organs other than the small intestine is consistent with previous reports (Pearson and McNulty, 1977; Theil et al, 1978; McAdaragh et a1, 1980). The virus-excretion patterns for pigs infected with the OSU and SDSU strains in the present study were similar and in agreement with previous reports (Lecce et a1, 1976; Bohl et a1, 1978; Tzipori and Williams, 1978; Torres-Medina and Underdahl, 1980). The greatest number of rotavirus particles as detected by electron microscopy were shed by gnotobiotic pigs at 72 hours PI. The number of rotavirus particles was reduced, or virus particles could not be detected 7 days after infection. As has been shown by other investigations (Torres—Medina and Underdahl, 1980), virus could be detected by electron microscopic examination of feces for a longer period of time when compared to immuno- fluorescence of the small intestine. Enterocytes infected by rotaviruses desquamate so immunofluorescent cells can only be detected up to 96 to 168 hours PI (Theil et a1, 1978; McAdaragh et a1, 1980). Immuno— fluorescent cells may be detected for even shorter periods of time after infection by some porcine rotavirus strains (Theil et al, 1985). Tl seroty; Dakota determi virus : lesion seroty} F inocul homoge the Oh Dakota rotavi Mortal monitc inocul and 16 the 3 killer c011e< immuH( eXamit SUMMARY The purpose of the present study was to compare serotypes 1 (Ohio State University strain) and 2 (South Dakota State University strain) of porcine rotavirus to determine if specific clinical signs, mortality rates, virus shedding patterns, virus antigen distribution, or lesion distribution and severity were related to the serotype of rotavirus with which pigs were infected. Forty hysterotomy derived gnotobiotic pigs were inoculated orally at 3 days of age with 2 ml of homogenate containing 105 pig-infectious dose50 of either the Ohio State University (OSU) strain or the South Dakota State University (SDSU) strain of porcine rotavirus. Controls were inoculated with media only. Mortality, clinical signs, and body weights were monitored daily. Five pigs in each of the virus inoculated groups and 4 control pigs were killed at 24 and 168 hours after inoculation. Four pigs in each of the 3 experimental groups (OSU, SDSU, and control) were killed 72 hours after inoculation. Specimens were collected at necropsy for serum chemistry, histologic, immunofluorescent, and scanning electron microscopic examinations. Al strains througf SDSU st hours t observ« OSU st did no strain but tt given immun< given after duode and i and f flexx comp; Alth. the atro 3-da Cont of} 117 All of the pigs inoculated with the OSU or SDSU strains survived. Control pigs remained healthy throughout the study. Pigs inoculated with the OSU or SDSU strains developed diarrhea 19-48 hours and 24—54 hours after inoculation, respectively. Vomiting was observed in 5 of 14 (362) of the pigs infected with the OSU strain whereas pigs inoculated with the SDSU strain did not vomit. Pigs inoculated with the OSU or SDSU strains had reduced weight gain compared to control pigs, but there was no difference in weight gain between pigs given the OSU or SDSU strains. Results of immunofluorescent examinations were similar for pigs given either rotavirus strain except that at 24 hours after inoculation, viral antigen was detected at the duodenal-jejunal flexure in 5/5 pigs given the OSU strain and in 1/5 pigs given the SDSU strain. Villous atrophy and fusion were more severe at the duodenal-jejunal flexure of pigs inoculated with the SDSU strain when compared to pigs inoculated with the OSU strain. Although the OSU and SDSU strains did show differences in the occurrence of vomiting and distribution of villous atrophy, these strains were equally virulent for 3—day-old gnotobiotic pigs. Therefore, prevention and control practices must be directed against both serotypes of porcine rotavirus. BIBLIOGRAPHY Acree modii bacte beef Adams the i of e; 51:35 Argex C.: gastt Bacht Kinet infEt Neon; l983 Banf: obse: mice 1968 Benf: Shed: farrt Benf- immu: anti and‘ 1984 Benj path. 71 Fish T o mneo east BIBLIOGRAPHY Acres, S. D., Radostits, 0. M.: The efficacy of a modified live reo-like virus vaccine and an E. coli bacterin for prevention of acute neonatal diarrhea of beef calves. Can Vet J 17:197-212, 1976. Adams, W. R., Kraft, L.: Electron microscopic study of the intestinal epithelium of mice infected with an agent of epizootic diarrhea of infant mice. Am J Pathol 51:39—60, 1967. Argenzio, R. A., Moon, H. W., Kemeny, L. J., Whipp, S. C.: Colonic compensation in transmissible gastroenteritis of swine. Gastroent 86:1504-1509, 1984. Bachmann, P. A., Hess, R. G., Dirksen, G., Schmid, C.: Kinetics of the local immune response to rotavirus infections in calves. Proceedings, 4th Internatl Symp Neonatal Diarrhea, Saskatoon, Sask, VIDO, pp 435-446, 3. Banfield, W. G., Kasnic, G., Blackwell, J. H.: Further observations on the virus of epizootic diarrhea of infant mice: an electron microscopic study. Virol 36:411-421, 1968. Benfield, D. A., Stotz, I., Moore, R., McAdaragh, J. P.: Shedding of rotavirus in feces of sows before and after farrowing. J Clin Micro 16:186—190, 1982. Benfield, D. A., Stotz, I. J., Nelson, E. A., Groon, K. 8.: Comparison of a commercial enzyme—linked immunosorbent assay with electron microscopy, fluorescent antibody, and virus isolation for the detection of bovine and porcine rotavirus. Am J Vet Res 45(10):l998—2002, 1984. Benjamin, M. H.: Outline of veterinary clinical pathology. The Iowa State University Press, Ames, IA, p 71, 1978. Bishop, R. P., Davidson, G. P., Holmes, I. N., Ruck, B. J.: Virus particles in epithelial cells of duodenal mucosal from children with acute non—bacterial gastroenteritis. Lancet ii:128l—1283, 1973. Bohl, Agnes diarr] Bohl, Cross diffei gnotol Bohl, serotj compai 19(2)‘ Bradbt R. G., syster rotav: Bridge partie 31:24f Bridge calf] Bridge porcit bovine Bridge Charat rotavi Bridge relatj rotan Bridge tYpice 116:5( Carpi( M.: I infect Synthe i’———i—iir¢:—i— 119 Bohl, E. H., Kohler, E. M. Saif, L. J. Cross, R. F., Agnes, A. G., Theil, K. W. Rotavirus as a caus of diarrhea in pigs. J Am Vet Med Assoc 172: 458- 463, 1978. Bohl, E. H., Saif, L. J., Theil, K. W., Agnes, A. G., Cross, R. F.: Porcine pararotavirus: detection, differentiation from rotavirus and pathogenesis in gnotobiotic pigs. J Clin Microbiol 15:312-319, 1982. Bohl, E. H., Theil, K. W., Saif, L. J.: Isolation and serotyping of porcine rotaviruses and antigenic comparison with other rotaviruses. J Clin Microbiol 19(2):105-111, 1984. Bradburne, A. F., Almeida, J. D., Gardner, P. S., Moosai, R G., Nash, A. A., Coombs, R. R. A.: A solid-phase system (space) for the detection and quantification of rotavirus in faeces. J Gen Virol 44:615-623, 1979. Bridger, J. C., Woode, G. N.: Characterization of two particle types of calf rotavirus. J Gen Virol 31:245-250, 1976 Bridger, J. D.: Location of type-specific antigens in calf rotaviruses. J Clin Microbiol 8(6):625—628, 1978. Bridger, J. C., Brown, J. F.: Development of immunity to porcine rotavirus in piglets protected from disease by bovine colostrum. Infect Immun 31(3):906-910, 1981. Bridger, J. C., Clark, I. N., McCrae, M. A.: Characterization of an antigenically distinct porcine rotavirus. Infect Immun 35:1058—1062, 1982. Bridger, J. C., Brown, J. F.: Antigenic and pathogenic relationships of three bovine rotaviruses and a porcine rotavirus. J Gen Virol 65:1151-1158, 1984. Bridger, J. D., Brown, J. F.: Prevalence of antibody to typical and atypical rotaviruses in pigs. Vet Rec 116:50, 1985. Carpio, M. M., Babiuk, L. A., Miska, V., Blumenthal, R. M.: Bovine rotavirus—cell interactions: effect of virus infection on cellular integrity and macromolecular synthesis. Virol 114:86-97, 1981a. Carpio, M., Bellamy, J. E. C., Babiuk, L. A.: Comparative virulence of different bovine rotavirus isolates. Can J Comp Med 45:38-42, 1981b. Castru Angeli cytopa Procee Saskat Chasey and it Virol Chase} experf 1977b Chase cattL Chrys False babie Clark Prodt J Cli Corne Chang with 2:10 Cohe asso 36:3 Conn foal Cort and .I If Cou] G. i rote str; Crm mul Pig ll: Cro cor Con 120 Castrucci, G., Ferrari, M., Frigeri, F., Cilli, V., Angelillo, G., Caleffi, F., Aldrovandi, V.: Studies on cytopathic strains of bovine rotavirus isolated in Italy. Proceedings, 4th International Symp Neonatal Diarrhea, Saskatoon, Sask, VIDO, pp 56-68, 1983. Chasey, D.: Different particle types in tissue culture and intestinal epithelium infected with rotavirus. J Gen Virol 37:443-451, 1977a. Chasey, D., Luc cas, M.: Detection of rotavirus in experimentally infected piglets. Res Vet Sci 22: 124-125, l977b. Chasey, D., Da avi : Atypical rotaviruses in pigs and cattle. Vet Rec e114: 16- 17, 1984. Chrystie, I. L., Totterdell, B. M., Banatuala, J. E.: False positive rotazyme tests on faecal samples from babies. Lancet ii:1028, 1983. Clark, S. M., Barnett, B. B., Spendlove, R. 8.: Production of high—titer bovine rotavirus with trypsin. J Clin Microbiol 9:413-417, 1979. Cornelius, L. M., Hooper, B. E., Haelterman, E. 0.: Changes in fluid and electrolyte balance in baby pigs with transmissible gastroenteritis. Am J Vet Clin Pathol 2:105—113, 1968. Cohen, J.: Ribonucleic acid polymerase activity associated with purified calf rotavirus. J Gen Virol 36:395-402, 1977. Conner, M. E., Darlington, R. W.: Rotavirus infection in foals. Am J Vet Res 41(10):l699-l703, 1980. Corthier, G., Vannier, P.: Production of coproantibodies and immune complexes in piglets infected with rotavirus. J Infect Dis l47(2):293—296, l983. Coulson, B. S., Fowler, K. J., Bishop, R. F., Cotton, R. G. H.: Neutralizing monoclonal antibodies to human rotavirus and indications of antigenic drift among strains of neonates. J Virol 54(1):l4—20, 1985. Crouch, C. F., Woode, G. N.: Serial studies of virus multiplication and intestinal damage in gnotobiotic piglets infected with rotavirus. J Med Microbiol 11:325—333, 1978. Crouch, C. F., Acres, S. D.: Prevalence of rotavirus and coronavirus antigens in the feces of normal cows. Can J Comp Med 48: 340— 342 , 1984. Davids Hamilt conven Drolet factor piglet virus. Duhame experf calves Confe: Chica Echev Vibul Rotav J Cli Elden T—cel exper 1986. E18, SA-ll 1972, EngL iden from 41:7 Engv assa Espe poly try; Este Rot; 43H Est enh mec Fer Nor a I out 197 121 Davidson, G. P., Gall, D. G., Petric, M., Butler, D. G., Hamilton, J. R.: Human rotavirus enteritis induced in conventional piglets. J Clin Invest 60:1402-1409, 1977. Drolet, R., Morin, M., Fontaine, M.: Hypoglycemia: a factor associated with low survival rate of neonatal piglets infected with transmissible gastroenteritis virus. Can J Comp Med 48:282—285, 1984. Duhamel, G. E., Osburn, B. I.: Cell mediated immunity to experimental bovine rotavirus infection in newborn calves. Proceedings, 66th Annual Meeting of the Conference of Research Workers on Animal Disease, Chicago, IL, p 49, 1985. Echeverria, P., Blacklow, N. R., Cukor, G. G., Vibulbandhitkit, S., Changchawalit, S., Bodnthai, P.: Rotavirus as a cause of severe gastroenteritis in adults. J Clin Microbiol 18:663-667, 1983. Elden, J., Leberman, H. M., Vonderfecht, S., Yolken, R.: T—cell-deficient mice display normal recovery from experimental rotavirus infection. J Virol 57:706—708, 986. E13, H. J., Lecatsas, G.: Morphology of the simian virus SA-ll and the "related" 0 agent. J Gen Virol 17:129-132, 1972. England, J. J., Poston, R. P.: Electron microscopic identification and subsequent isolation of a rotavirus from a dog with fatal neonatal diarrhea. Am J Vet Res 41:782-783, 1980. Engvall, E., Perlmann, P.: Enzyme-linked immunosorbent assay, ELISA. J Immunol 109:129-135, 1972. Espejo, R. T., Lopez, 5., Arias, C.: Structural polypeptides of simian rotavirus SA—ll and the effect of trypsin. J Virol 37:156-160, 1981. Estes, M. K., Grahman, D. Y., Smith, E. M., Gerba, C. P.: Rotavirus stability and inactiviation. J Gen Virol 43:403-409, 1979. Estes, M. K., Graham, D. Y., Mason, B. B.: Proteolytic enhancement of rotavirus infectivity: molecular mechanisms. J Virol 39:879—889, 1981. Fernelius, A. L., Ritchie, A. E., Classick, L. B., Norman, J. 0., Mebus, C. A.: Cell culture adaptation of a reovirus-like agent of calf diarrhea from a field outbreak in Nebraska. Arch Ges Virusforsh 37:114—130, 1972. Flewe Bridg from calve Flewe diarr 1975. Flewe Virol Flewe infec 19781 Ganor funct Medic Gethi Clon: hemag antn Natu: Grah; Pathe stud 29:U Grah clas L'In Gree W') none duri bovi 420— Gree K‘) Flor Char Surf 47:2 Hall N.: neor Vet 122 Flewett, T. H., Bryden, A. 8., Davies, H., Woode, G. N., Bridger, J. C. , Derrick, J. M. Relation between viruses from acute gastroenteritis of children and newborn calves. Lancet ii: 61- 63, 4. Flewett, T. H., Bryden, A. 8., Davies, H.: Virus diarrhoea in foals and other animals. Vet Rec 96:477, 1975. Flewett, T. H., Woode, G. N.: The rotaviruses. Arch Virol 57:1—23, l978a. Flewett, T. H.: Electron microscopy in the diagnosis of infectious diarrhea. J Am Vet Med Assoc 173:538—543, 1978b. Ganong, W. F.: Neural centers regulating visceral function. In: Review of Medical Physiology, Lan ge Medical Publications, Los Altos, CA, pp 178—179, 1983. Gething, M. J., Bye, J., Skehel, J., Waterfield, M.: Cloning and DNA sequence of double—stranded copies of hemagglutinin genes from H2 and H3 strains elucidates antigenic shift and drift in human influenza virus. Nature 287:301-306, 1980. Graham, D. Y. Sackman, J. W. Estes, M. R. : Pathogenesis of rotavirus- -induced diarrhea. preliminary studies in miniature swine piglet. Dig Dis Sci 29:1028—1035, 1984. Graham, D. Y., Estes, M. K.: Proposed working serologic classification system for rotaviruses. Annales de L'Institut Pasteur/de Virologie 136:5—12, 1985. Greenberg, H. B., Kalica, A. R., Wyatt, R. G., Jones, R. W., Kapikian, A. Z., Chanock, R. M.: Rescue of noncultivatable human rotavirus by gene reassortment during mixed infection with ts mutants of a cultivatable bovine rotavirus. Proceedings, Natl Acad Sci, USA, pp 420—424, 1981. Greenberg, H. B., Valdesuso, J., Vansyke, K., Midthum, K., Walsh, M., McAulife, V., Wyatt, R. G., Kalica, A. R., Flores, J., Hoshino, Y.: Production and preliminary characterization of monoclonal antibodies directed at two surface proteins of rhesus rotavirus. J Virol 47. 267— 275, 1983. Hall, G. A., Bridger, J. C., Chandler, R. L., Woode, G. N.: Gnotobiotic piglets experimentally infected with neonatal calf diarrhoea reovirus—like agent (rotavirus). Vet Pathol 13:197-210, 1976. Hess, to rot infec 42:1L Hoble study Annua Anima Hoope patho gastr 149:1 Hoshi Chara 54:31 Hoshi 123 Hess, R. G., Bachmann, P. A.: Distribution of antibodies to rotavirus in serum and lacteal secretions of naturally infected swine and their suckling pigs. Am J Vet Res 42:1149-1152, 1981. Hoblet, K. H., Saif, L. J., Kohler, E. M. An efficacy study of a porcine rotavirus vaccine. Proceedings, 65th Annual Meeting of the Conference of Research Workers in Animal Disease, Chicago, IL, p 53,4. Hooper, B. E., Haeltermann, E. 0.: Concepts of pathogenesis and passive immunity in transmissible gastroenteritis of swine. J Am Vet Med Assoc 149:1580—1586, 1966. Hoshino, Y., Baldwin, C. A., Scott, F. W.: Isolation and characterization of feline rotavirus. J Gen Virol 54:313-323, 1981. Hoshino, Y., Wyatt, R. G., Greenberg, H. B., Flores, J., Kapikian, A. Z.: Serotypic similarity and diversity of rotaviruses of mammalian and avian origin as studied by plaque-reduction neutralization. J Infect Dis 149:694—702, 1984 Huffman, E. M., Clark, C. H., Olson, J. D.: Serum chloramphenicol concentrations of preruminant calves: A comparison of two formulations dosed orally. J Vet Pharmacol Ther 4:225-231, 1981. Johnson, C. A., Fulton, R. W., Henk, W. G., Snider, T. G.~ Inoculation of neonatal gnotobiotic dogs with a canine rotavirus. Am J Vet Res 44:1682—1693, 1983. Kalica, A. R., Greenberg, H. B., Wyatt, R. G., Flores, J., Sereno, M. M., Kapikian, A. Z., Chanock, R. M: Genes of human (strain Wa) and bovine (strain UK) rotaviruses that code for neutralization and subgroup antigens. Virol 112:385~390, 1981a. Kalica, A. R., Greenberg, H. B., Espejo, R. T., Flores, J., Wyatt, R. G., Kapikian, A. Z., Chanock, R. M.: Distinctive ribonucleic acid patterns of human rotavirus subgroups l and 2. Infect Immun 33:958-961, 1981b. Kapikian, A. Z., Chanock, R. M.: Rotaviruses. In: Virolo (B. N. Fields, ed), Raven Press, New York, p 7 , 985. Kapikian, A. Z., Cline, W. L., Mebus, C. A., Wyatt, R. G., Kalica, A. R., James, H. D., Vankirk, D., Chanock, R. M., Kimb, H. W.: New complement—fixation test for the human reovirus—like agent of infantile gastroenteritis. Lancet 1:1056—1061, 1975. Kapiki Chanoc agent Med 29 Kapiki R. G., Flores of but Karnox of hig Cell] Kerznt Tranm intes enter Lecce assoc Immun Lecce (rota for r 43:9C Lecce infee wean: Intel PF 3' Litt Patt and Pr0c Sask Lour Bric Elec isol Make ' ’ CeL 124 Kapikian, A. Z., Kim, H. W., Wyatt, R. G., Cline, W. L., Chanock, R. M., Parrott, R. H.: Human reovirus-like agent associated with "winter" gastroenteritis. N Engl J Med 294:965-972, 1976. Kapikian, A. Z., Cline, W. L., Greenberg, H. B., Wyatt, R G., Kalica, A. R., Banks, C. E., James Jr., H. D., Flores, J., Chanock, R. M.: Antigenic characterization of human and animal rotaviruses by immune adherence hemagglutination assay (IAHA): evidence for distinctness of IAHA and neutralization antigens. Infect Immun 33:415-425, 1981. Karnovsky, M. J.: A formaldehyde-glutara1dehyde fixative of high osmolarity for use in electron microscopy. Cell Biol 27:137A, 1965. Kerzner, B., Kelly, M. H., Gall, D. G., Butler, D. C.: Transmissible gastroenteritis: sodium transport and the intestinal epithelium during the course of viral enteritis. Gastroent 72:457—461, 1977. Lecce, J. G., King, M. W., Mock, R.: Reovirus—like agent associated with fatal diarrhea in neonatal pigs. Infect Immun 14:816—825, 1976. Lecce, J. G., King., M. W.: The calf reo-like virus (rotavirus) vaccine: an ineffective immunization agent for rotaviral diarrhea of piglets. Can J Comp Med 43:90-93, 1979. Lecce, J. C., King, M. W.: Persistent rotaviral infection producing multiple episodes of diarrhea in weanling pigs reared in isolation. Proceedings, 3rd Internatl Symp Neonatal Diarrhea, Saskatoon, Sask, VIDO, pp 27-36, 1980. Little, L. M., Shadduck, J. A., Gelberg, H. B., Patterson, J. 8.: Variables responsible for induction and termination of rotavirus diarrhea in mice. Proceedings, 4th Internatl Symp Neonatal Diarrhea, Saskatoon, Sask, VIDO, pp 69-74 1983. Lourenco, M. H., Nicolas, J. C., Cohen, J., Scherrer, R., Bricout, F.: Study of human rotavirus genome by . electrophoresis: attempt of classification among stra1ns isolated in France. Ann Virol 132:161-173, 1981. Makabe, T., Komaniwa, H., Kishi, Y., Yataya, K., Imagawa, H., Sato, K., Inaba, Y.: Isolation of ovine rotaV1rus 1n cell cultures. Arch Virol 83:123-127, 1985. Malhe SA-ll 22:23 Malic thioc micro exper Marti Ultra Virol Maset dehye micre Cyto< Math; subut Viro Matt‘ of v Mavr A. S in r D-xy McAd Johr R.: pigs 41(1 McNt D.: aSSt 1:5.| MCN‘ J_' age 197 McN cul Vir MCF inf 125 Malherbe, H. H., Strickland-Cholmley, M.: Simian virus SA—ll and the related 0 agent. Arch ges Virusforsch 22:235-245, 1967. Malick, L. E., Wilson, R. B.: Modified thiocarbohydrazide procedure for scanning electron microscopy: routine use for normal, pathological, or experimental tissues. Stain Tech 50:265-269, 1975. Martin, M. L., Palmer, E. L., Middleton, P. J.: Ultrastructure of infantile gastroenteritis virus. Virology 68:146-153, 1975. Maser, M. H., Trimble, J. J.: Rapid chemical dehydration of biological samples for scanning electron microscopy using 2, 2,—dimethoxypropane. J Histochem Cytochem 25:247-251, 1977. Mathan, M., Almeida, J. D., Cole, J.: An antigenic subunit present in rotavirus infected faeces. J Gen Virol 34:325-329, 1977. Matthews, R. E. F.: The classification and nomenclature of viruses. Intervirol 11:133-135, 1979. Mavromichaelis, J., Evans, N., McNeish, A. S., Bryden, A. S., Davies, H. A., Flewett, T. H.: Intestinal damage in rotavirus and adenovirus gastroenteritis assessed b D-xylose malabsorption. Arch Dis Child 52:589-591, 1977. McAdaragh, J. P., Bergeland, M. E., Meyer, R. C., Johnshoy, M. W., Stotz, I. J., Benfield, D. A., Hammer, Ri: Pathogenesis of rotaviral enteritis in gnotobiotic s: a microscopic study. Am J Vet Res 41(10): 1572-1580, 1980. McNulty, M. S., Pearson, G. R., McFerran, J. B., Collins, D. S., Allan, G. M.: A reovirus-like agent (rotavirus) associated with diarrhea in neonatal pigs. Vet Microbiol 1:55-63, 1976a. McNulty, M. S., Allan, G. M., Pearson, G. R., McFerran, J. B., Curran, W. L., McCracken, R. M.: Reovirus-like agent (rotavirus) from lambs. Infect Immun 14:1332-1338, 1976b. McNulty, M. S., Allan, G. M., McFerran, J. B.: Cell culture studies with a cytopathic bovine rotavirus. Arch Virol 54:207-209, 1977. McNulty, M. S., Allan, G. M. , Stuart, C. : Rotavirus infection in avian species. Vet Rec 103: 319— 320, 1978. McNul 13013 from McNul Exper clini l983. Mebus from 1969. Mebus J.: (sc01 Mebu: reoé Mebu Immu Asso Mebu lesi huma 1977 Midd Bort gast Midd PrOp grou Immt Midc for (0rt 29:] Mid( J Ar Midt I‘Ote Cant 126 McNulty, M. S., Allan, G. M., Todd, D., McFerran, J. B.: Isolation and cell culture propagation of rotaviruses from turkeys and chickens. Arch Virol 61:13—21, 1979. McNulty, M. S., Allan, G. M., McCracken, R. M.: Experimental infection of chickens with rotaviruses: clinical and virologic findings. Avian Pathol 12:45—54, 1983. Mebus, C. A., Underdahl, N. R., Rhodes, M. B., Twiehaus, M. J.: Calf diarrhea (scours): reproduced with a virus from a field outbreak. Res Bull Univ Nebraska, 233, 1969 Mebus, C. A., Kono, M., Underdahl, N. R., Twiehaus, M. J.: Cell culture propagation of neonatal calf diarrhea (scours) virus. Can Vet J 12:69-72, 1971a. Mebus, C. A., Stair, E. L., Underdahl, N. R., Twiehaus, M. J.: Pathology of neonatal calf diarrhea induced by a reo-like virus. Vet Pathol 8:490—505, 1971b. Mebus, C. A., White, R. G., Bass, E. P., Twiehaus, M. J.: Immunity to neonatal calf diarrhea Virus. J Am Vet Med Assoc 163:880-883, 1973. Mebus, C. A., Wyatt, R. G., Kapikian, A. Z.: Intestinal lesions induced in gnotobiotic calves by the virus of human infantile gastroenteritis. Vet Path 14:273—282, 977. Middleton, P. J., Szymanski, M. T., Abbott, G. D., Bortolussi, R., Hamilton, J. R.: Orbivirus acute gastroenteritis of infancy. Lancet 1:1241—1244, 1974. Middleton, P. J., Petric, M., Szymanski, M. T.: Propagation of infantile gastroenteritis virus (Orbi group) in conventional and germ-free piglets. Infect Immun 12:1276-1280, 1975. Middleton, P. J., Petric, M., Hewitt, C. M., Szymanski, M. T., Tam, J. S.: Counter-immunoelectro—osmophoresis for the detection of infantile gastroenteritis virus (Orbi group) antigen and antibody. J Clin Pathol 29:191—197, 1976. Middleton, P. J.: Pathogenesis of rotaviral infection. J Am Vet Med Assoc 1731544-545, 1978. Midthun, K., Greenberg, H. B., Hoshino, Y., Kapikian, A. Z., Wyatt, R. G., Chanock, R. M.: Reassortant rotaviruses as potential live rotavirus vaccine candidates. J Virol 53:949~954, 1985. Minie and 1 Moon, mucoe Med 1 Moon, Boot} gasti cell intes Morn Comp: in m l‘lOUWt Ster Veti Mouw Kijk in p Mura Sero neut Nari Inte inoc Q(J News Esch 1.x. .mmr Offi Mole segu Offj Bacl anti pre; 20:: Off: rote Pas: ant: 127 Miniats, O. P., Jol, D.: Gnotobiotic pigs-—derivation and rearing. Can J Comp Med 42:428—437, 1978. Moon, H. W.: Epithelial cell migration in the alimentary mucosa of the suckling pig. Proceedings, Soc Exp Biol Med 13:151, 1971. Moon, H. W., Kemeny, L. J., Lambert, G., Stark, S. L., Booth, G. D.: Age dependent resistance to transmissible gastroenteritis of swine. III. Effects of epithelial cell kinetics on coronavirus production and on atrophy of intestinal villi. Vet Path 12:434—445, 1975. Morinet, F., Ferchal, F., Colimon, R., Perol, Y.: Comparison of six methods for detecting human rotavirus in stools. Fur J Clin Microbiol 3:136-140, 1984. Mouwen, J. M. V. M.: White scours in piglets. I. Steriomicroscopy of the mucosa of the small intestine. Vet Pathol 8:364—380, 1971. Mouwen, J. M. V. M., Shotman, A. J. H., Wensing, T., Kijkuit, C. J.: Some biochemical aspects of white scours in piglets. Tijdschr Diergeneesk 97:65—90, 1972. Murakami, Y., Nishioka, N., Hashiguchi, Y., Kuniyasu, C.: Serotypes of bovine rotaviruses distinguished by serum neutralization. Infect Immun 40:851-855, 1983. Narita, M., Fukusho, A., Konno, S., Shimizu, Y.: Intestinal changes in gnotobiotic piglets experimentally inoculated with porcine rotavirus. Natl Inst Anim Health Q (JPN) 22:54-60, 1982. Newsome, P. M., Coney, K. A.: Synergistic rotavirus and Escherichia coli diarrheal infection of mice. Infect Immun 42:573—574, 1985. Offit, P. A., Blavat, G., Greenberg, H. B., Clark, H. F.: Molecular basis of rotavirus virulence: Role of gene segment 4. J Virol 57:46-49, 1986. Offit, P. A., Clark, H. F., Taylor, A. H., Hess, R. G., Bachmann, P. A., Plotkin, S. A.: Rotavirus—specific antibodies in fetal bovine serum and commercial preparations of serum albumin. J Clin Microbiol 20:266-270, 1984. Offit, P. A., Clark, H. F.: Protection against rotavirus-induced gastroenteritis in a murine model by passively acquired gastrointestinal but not circulating antibodies. J Virol 54:58—64, 1985. Palme and s compa Virol Panel disea 173:5 Pears the e reovi 1977. Pears in SD with Pedle Molee grout PedL Defh Gen‘ Petr Hewi epiz 1978 Petr lden Inte Petr Loca ultr 63:4 Quan the lnte Rebe trar Pigs Reyy C.: dete Vet 128 Palmer, E. L., Martin, M. L., Murphy, F. A.: Morphology and stability of infantile gastroenteritis virus: comparison with reovirus and bluetongue virus. J Gen Virol 35:403-414, 1977. Panel report on the colloquium on selected diarrheal diseases of the young. J Am Vet Med Assoc 173:515-518, 1978. Pearson, G. R., McNulty, M. S.: Pathological changes in the small intestine of neonatal pigs infected with a pig reovirus-like agent (rotavirus). J Comp Path 87:363—375, 1977. Pearson, G. R., McNulty, M. S.: Ultrastructural changes in small intestinal epithelium of neonatal pigs infected with pig rotavirus. Arch Virol 59:127-136, 1979. Pedley, S., Bridger, J. C., Brown, J. F., McCrae, M. A.: Molecular characterization of rotaviruses with distinct group antigens. Virol 64:2093-2101, 1983. Pedley, S., Bridger, J. C., Chasey, D., McCrate, M. A.: Definition of two new groups of atypical rotaviruses. J Gen Virol 67:131—137, 1986. Petric, M., Middleton, P. J., Grant, C., Tam, J. S, Hewitt, C. M.: Lapine rotavirus-—preliminary studies on epizootiology and transmission. Can Com Med 42:143-147, 1978. Petrie, B. L., Graham, D. Y., Estes, M. K.: Identification of rotaviral particle types. Intervirology 16:20—28, 1981. Petrie, B. L., Graham, D. Y., Hanssen, H., Estes, M. K.: Localization of rotavirus antigens in infected cells by ultrastructural immmunocytochemistry. J Gen Virol 63:457-467, 1982. Quan, C. M., Doane, F. W.: Ultrastructural evidence for the cellular uptake of rotavirus by endocytosis. Intervirology 20:223-231, 1983. Reber, E. F., Whitehair, C. K.: The effect of transmissible gastroenteritis on the metabolism of baby pigs. Am J Vet Res 16:116—119, 1955. Reynolds, D. J., Chasey, D., Scott, A. C., Bridger, J. C.: Evaluation of ELISA and electron microscopy for the detection of coronavirus and rotavirus in bovine faeces. Vet Rec 114:397—401, 1984. Rey R.: cln Rim Bar: rott Bioi Rie] Chai rote Symt Stat IreI Rite mic1 virt Rodg Bioe virt 1975 Rodg of e infe Rodr mice M01 Rodr D" Parr gast in i 1977 Rodr K'! huma invo Infe Rube eyzy for 1982 129 Reynolds, D. J., Hall, G. A., Debney, T. G., Parsons, K. R.: Pathology of natural rotavirus infection in clinically normal calves. Res Vet Sci 38:264-269, 1985. Riepenhoff—Talty, M., Lee, P. C., Carmody, P. J., Barrett, H. J., Ogra, P. L.: Age-dependent rotavirus-enterocyte interactions. Proceedings, Soc Exp Biol Med 170:146-154, 1982a. Riepenhoff-Talty, M., Suzuki, H., Ogra, P. L.: Characteristics of the cell-mediated immune response to rotavirus in suckling mice. Proceedings, International Symposium on Enteric Infections in Man and Animals: Standardization of Immunological Procedures, Dublin, Ireland, Develop Biol Standard, pp 263—268, 1982b. Ritchie, A. E., Fernelius, A. L.: Direct immune—electron microscopy and some morphological features of hog cholera virus. Arch Gesamte Virus-forsch 23:292-298, 1968. Rodger, S. M., Schnagl, R. G., Holmes, I. H.: Biochemical and biophysical characteristics of diarrhea viruses of human and calf origin. J Virol 16:1229-1235, 1975. Rodger, S. M., Bishop, R. F., Holmes, I. H.: Detection of a rotavirus-like agent associated with diarrhea in an infant. J Clin Microbiol 16:724—726, 1982. Rodriguez-Tore, G.: Natural epizootic diarrhea of infant mice (EDIM) a light and electron microscope study. Exp Mol Path 32:241-252, 1980. Rodriguez, W. J., Kim, H. W., Arrobio, J. 0., Brandt, C. D., Chanock, R. M., Kapikian, A. Z., Wyatt, R. G. Parrott, R. H.: Clinical features of acute gastroenteritis associated with human reovirus-like agent in infants and young children. J Pediatr 91:188-193, 1977. ’ Rodriguez, W. J., Kim H. W., Brandt, C. D. , Gardner, M. K., Parott, R. H.: Use of electrophoresis of RNA from human rotavirus to establish the identity of strain involved in outbreaks in a tertiary care nursery. J Infect Dis 148:34-40, 1983. Rubenstein, A. S., Miller, M. F.: Comparison of an eyzyme immunoassay with electron microscopic procedures for detecting rotavirus. J Clin Microbiol 15:938—944, 1982. Rubi viru 1980 Saif Immr gast of s Sail port inte 1971 Sat Hou: 234 you Sai Pas fed cow Sai The pre Res Sai in 130 Rubin, D. H., Fields, B. N.: Molecular basis of reovirus virulence: Role of the M2 gene. J Exp Med 152:853—868, 1980. Saif, L. J., Bohl, E. H., Kohler, E. M., Hughes, J. H.: Immune electron microscopy of transmissible gastroenteritis virus and rotavirus (reovirus-like agent) of swine. Am J Vet Res 38:13-20, 1977. Saif, L. J., Theil, K. W., Bohl, E. H.: Morphgensis of porcine rotavirus in porcine kidney cell cultures and intestinal epithelial cells. J Gen Virol 39:205-217, 1978. Saif, L. J., Bohl, E. H., Theil, K. W., Cross, R. F., House, J. A.: Rotavirus-like, calicivirus—like, and 23—nm virus—like particles associated with diarrhea in young pigs. J Clin Microbiol 12:105-111, 1980. Saif, L. J., Redman, D. R., Smith, K. L., Theil, K. W.: Passive immunity to bovine rotavirus in newborn calves fed colostrum supplements from immunized or nonimmunized cows. Infect Immun 41:1118-1131, 1983. Saif, L. J., Smith, K. L., Landmeier, B. J., Bohl, E. H., Theil, K. W., Todhunter, D. A.: Immune response of pregnant cows to bovine rotavirus immunization. Am J Vet Res 45:49-58, 1984. Saif, L. J., Saif, Y. M., Theil, K. W.: Enteric viruses in diarrheic turkey poults. Avian Dis 29:798—811, 1985. Samplson, J., Hester, H. R., Graham, R.: Studies on baby pig mortality. II. Further observations on acute hypoglycemia in newly born pigs (so-called baby pig disease). J Am Vet Med Assoc 100:33—37, 1942. Sato, K., Inaba, Y., Shinozaki, T., Matumoto, M.: Neutralizing antibody to bovine rotavirus in various animal species. Vet. Microbiol 6:259—261, 1981 Sato, K., Inaba, Y., Shinozaki, T., Matumoto, M.: Isolation of human rotavirus in cell cultures. Arch Virol 71:267—271, 1982a. Sato, K., Inaba, Y., Miura, Y., Tokuhisa, S., Matumoto, M. : Antigenic relationships between rotaviruses from different species as studied by neutralization and immunofluorescence. Arch Virol 73: 45- 50, 1982b. Sattar, S. A., Raphael, R. A., Spring-Thorpe, V. S.: Rotaviruses and chemical disinfectants. Proceedings, 4th Internatl Symp Neonatal Diarrhea, Saskatoon, Sask, VIDO, pp 90—98, 1983. Satt Sprz‘ airl Mic1 Shi] Flet stal Sno< dis: SHOt rot; 197: Sno rot. Virt Sno for Spe 39: Sta int Pat Sta pha chi Sta mor 197 Str A ant tee Suz 1st hum 19E 131 Sattar, S. A., Ijaz, M. K., Johnson- -Lussenburg, C. M. Spring-Thorpe, V. S.: Effect of relative humidity on the airborne survival of rotavirus SA—11.App1 Environ Microbiol 47:879-881, 1984 Shirley, J. A., Beards, G. M., Thouless, M. E., F1ewett,T. H.: The influence of divalent cations on the stability of human rotavirus. Arch Virol 67:1-9, 1981. Snodgrass, D. R., Herring, J. A.: The action of disinfectants of lamb rotavirus. Vet Rec 101:81, 1977. Snodgrass, D. R., Wells, P. W.: Passive immunity in rotaviral infections. J Am Vet Med Assoc 173:565-568, 1978. Snodgrass, D. R. Herring, A. J., Campbell, I., Inglis, J. M., Hargreaves, F. D.: Comparison of atypical rotaviruses from calves, piglets, lambs, and man. J Gen Virol 65:909-914, 1984a. Snodgrass, D. R., Ojeh, C. K., Campbell, I., Herring, A. J.: Bovine rotavirus serotypes and their significance for immunization. J Clin Microbiol 20:342—346, 1984b. Spencer, E. G., Avendano, L. F., Garcia, B. 1.: Analysis of human rotavirus mixed electropherotypes. Infect Immun 39:569-574, 1983. Stair, E. L., Mebus, C. A., Twiehaus, M. J., Underdahl, N. R.: Neonatal calf diarrhea. Electron microscopy of intestines infected with a reovirus-like agent. Vet Pathol 10:155—170, 1973. Stals, F., Walther, F. J., Bruggeman, C. A.: Faecal and pharyngeal shedding of rotavirus and rotavirus IgA in children with diarrhoea. J Med Virol 14:333—339, 1984. Stannard, L. M., Schoub, B. D.: Observations n the morphology of two rotaviruses. J Gen Virol 37:435—439, 1977. Struker, G., Schmidt, N. J., Forghan, B., Holmberg, C. A., Henrickson, R. V., Anderson, J. H.: Rotavirus antibody assays on monkey sera: a comparison of enzyme immunoassay with neutralization and complement—fixation tests. Am J Vet Res 40:1620—1623, 1979. Suzuki, H., Konno, T., Kitaoka, S., Sato, T., Ebina, T. ! Ishida, N.: Further observations on the morphogenesis of human rotavirus in MA—104 cells. Arch Virol 79:147—159, 1984. Suzul Two I Arch Tajit Shine in C! T8111 Hami feat ch11 Thei PrOP Thei Agne infe pigl Thei of e Res Thei rote ant: Tho Sp]: Thu tri Vac Tor Esc Vet T01 mie Wit Tz 132 Suzuki, H., Kitaoka, S., Konno, T., Sato, T., Ishida, N.: Two modes of human rotavirus entry into MA-104 cells. Arch Virol 85:25—34, 1985. Tajima, T., Suzuki, E., Ushijima, H., Araki, K., Kim, B., Shinozaki, T., Fujii, R.: Isolation of murine rotavirus in cell cultures. Arch Virol 82:119—123, 1984. Tallett, S., MacKenzie, C., Middleton, P., Kerzner, B., Hamilton, R.: Clinical, laboratory, and epidemiological features of a viral gastroenteritis in infants and children. Pediatrics 60:217-222, 1977. Theil, K. W., Bohl, E H., Agnes, A. G.: Cell culture propagation of porcine rotavirus (reovirus-like agent). Am J Vet Res 38:1765-1768, 1977. Theil, K. W., Bohl, E. H., Cross, R. F., Kohler, E. M., Agnes, A. G.: Pathogenesis of porcine rotaviral infection in experimentally inoculated gnotobiotic piglets. Am J Vet Res 39:213—220, 1978. Theil, K. W., Bohl, E. H.: Porcine rotaviral infection of cell culture: effects of certain enzymes. Am J Vet Res 41:140-143, 1980. Theil, K. W., Saif, L. J.: In vitro detection of porcine rotavirus-like virus (group B rotavirus) and its antibody. J Clin Microbiol 21:844, 846, 1985a. Theil, K. W., Saif, L. J., Moorehead, P. D., Whitmoyer, R. E.: Porcine rotavirus—like virus (group B rotavirus): characterization and pathogenicity for gnotobiotic pigs. J Clin Microbiol 21:340-345, 1985b. Thompson, S. W., Hunt, R. D.: Selected Histochemical and Histopathological Methods. Charles C. Thomas, Springfield, IL, 1966. Thurber, E. T., Bass, E. P., Beckenahuer, W. H.: Field trial evaluation of a reo-coronavirus calf diarrhea vaccine. Can J Comp Med 41:131—136, 1977. Torres—Medina, A.: Effect of combined rotavirus and Escherichia coli in neonatal gnotobiotic calves. Am J Vet Res 45:643-660, 1984. Torres—Medina, A., Underdahl, N. R.: Scanning electron microscopy of intestine of gnotobiotic piglets infected with porcine rotavirus. Can J Comp Med 44:403-411, 1980. Tzipori, S., Caple, I. W, Butler, R.: Isolation of rotavirus from deer. Vet Rec 99:398, 1976. Tzipor: inocul Tzipor respon inocul isolat Tzipor Escher four-v Austre Uhnoo. featu: with l 23:55 Utera infec Vairn Immu: milk estir 1970 Van anti rota rota 1986 Vesi Zise atte adul VOHt c. , pro 198 War of Mic Wei rot jut 133 Tzipori, S., Williams, I. H.: Diarrhoea in piglets inoculated with rotavirus. Aust Vet J 54:188—192, 1978. Tzipori, S. R., Marin, T. J., Smith, M. L.: The clinical response of gnotobiotic calves, pigs, and lambs to inoculation with human calf, pig, and foal rotavirus isolates. Ajebak 58:309-318, 1980a. Tzipori, S., Chandler, D., Makin, T., Smith, M.: Escherichia coli and rotavirus infections in four—weeE-old gnotobiotic piglets fed milk or dry food. Austral Vet J 56:279-284, 1980b. Uhnoo, I., Svensson, L.: Clinical and epidemiological features of acute infantile gastroenteritis associated with human rotavirus subgroups 1 and 2. J Clin Microbiol 23:551-555, 1986. Utera, V., Mazzali de Ilja, R., Gorziglia, M., Esparza, J.: Epidemiological aspects of porcine rotavirus infection in Venezuela. Res Vet Sci 36:310-315, 1984. Vairman, J. P., Arbuckle, J. B., Heremans, J. F.: Immunoglobulin A in the pig. II. Sow's colostral and milk IgA: quantitative studies and molecular size estimation. Int Arch Allergy Appl Immunol 39:323—333, 1970. Van Zaane, D., Ijzerman, J., DeLeeuw, P. W.: Intestinal antibody response after vaccination and infection with rotavirus of calves fed colostrum with or without rotavirus antibody. Vet Immuno Immunopath 11:45-63, 1986. Vesikari, T., Isolauri, E., Delem, A., D‘Hondt, E., Zissis, G.: Immunogenicity and safety of live oral attenuated bovine rotavirus vaccine strain RIT 4237 in adults and young children. Lancet 11:808-811, 1983. Vonderfecht, S., L., Huber, A. C., Eiden, J., Mader, L. C., Yolken, R. H.: Infectious diarrhea of infant rats produced by a rotavirus—like agent. J Virol 52:94—98, 1984. Ward, R. L., Knowlton, D. R., Pierce, M. J.: Efficiency of human rotavirus propagation in cell culture. J Clin Microbiol 19:748-753, 1984. Weiss, C., Clark, H. F.: Rapid inactivation of rotaviruses by exposure to acid buffer or acid gastric juice. J Gen Virol 66:2725—2730, 1985. [1| 1| Wt Re 11 Sci. 134 Welch, A. B., Twiehaus, M. J.: Cell culture studies of a neonatal calf diarrhea virus. Can J Comp Med 37:287-294, 1973. Wenman, W. M., Hinde, D., Feltham, S. Gurwi th, Rotavirus infection in adult. N Engl J Med 301: 303— 306, 1979 White, L., Perez, I., Perez, M., Urbina, G., Greenberg, H., Kapikian, A., Flores, J.: Relative frequency of rotavirus subgroups l and 2 in Venezuelan children with gastroenteritis as assayed with monoclonal antibodies. J Clin Microbiol 19:516-520, 1984. Woode, G. N. Bridger, J. C. Viral enteritis of calves. Vet Rec 96: 85— 88, 1975a. Woode, G. N.: Pathogenic rotaviruses isolated from pigs and calves. In: Acute Diarrhoea in Childhood, CIBA Found Symp 42:251-271, 1975b. Woode, G. N., Bridger, J. C., Jones, J. M., Flewett, T. H., Bryden, A. S., Davies, H. A., White, G. B. B.: Morphological and and antigenic relationships between viruses (rotaviruses) from acute gastroenteritis of children, calves, piglets, mice, and foals. Infect Immun 41:804—810, 1976a. Woode, G. N., Bridger, J., Hall, G. A., Jones, J. M., Jackson, G. The isolation of reovirus—like agents (rotavirus) from acute gastroenteritis of piglets. J Med Microbiol 9:203-209, 1976b. Woode, G. N., Smith, C., Dennis, M. J.: Intestinal damage in rotavirus infected calves assessed by D-xylose malabsorption. Vet Rec 102:340-341, 1978. Woode, G. E. Viral infections of the intestinal tract: Pathological and clinical aspects. In: Viral Enteritis in Humans and Animals. Inserm Symp Ser 90 0:15-38 I97 9. Woode, G. N., Bohl, E. H.: Porcine rotavirus infections. In: Disease of Swine, Leman et al, eds, Iowa State Univ Press, Ames, Iowa, pp 310—322, 1981. Wyatt, R. G., Mebus, C. A. Yolken, R. H., Kalica, A. R., James Jr., H. D,. Kapikian, A. Z., Chanocik, R. M.: Rotaviral immunity in gnotobiotic calves: Heterologous resistance to human virus induced by bovine virus. Science 203:5448-550, 1979. Stra 135 Wyatt, R. G. James, W. D., Bohl, E. H., Thiel, K. W., Saif, L. J., Kalica, A. R. Greenberg, H. B., Kapikian, A. Z. , Chanock, R. M.: Human rotavirus type : cultivation in vitro. Science 207: 189-191, 1980. Wyatt, R. G., James, W. B: Methods of gastroenteritis virus culture in vivo and in vitro. In: Virus Infections of the Gastrointestinal Tract, (D. J. Tyrell and A. Z. Kapikian, eds.), MarceliDekker, Inc., New York, pp 13-35, 1982. Yamaguchi, H., Inouye, S., Yamauchi, M., Morishima, T., Matsuno, S., Isomusa, S., Suzukie, S.: Anamestic response in fecal IgA antibody production after rotaviral infection of infants. J Infect Dis 152:398-399, 1985. Yolken, R. H., Barbour, B., Wyatt, R. G., Kalica, A. R., Kapikian, A. Z., Chaock, R. M.: Enzyme-linked immunosorbent assay for identification of rotaviruses from different animal species. Science 201:254, l978a. Yolken R. H., Wyatt, R. G., Zissis, G., Brandt, C. D., Rodriguez, W. J., Kim, H. W., Parrott, R. H., Urrutia, J. J., Mata, L., Greenberg, H. B., Kapikian, A. Z., Chanock, R. M.: Epidemiology of human rotavirus types 1 and 2 as studied by enzyme-linked immunosorbent assay. N Engl J Med 299:1156-1161, l978b. Yolken, R. H., Wyatt, R. G., Barbour, B. A., Kim, H. W., Kapikian, A. Z., Chanock, R. M.: Measurement of anti-rotavirus antibody by an enzyme immunosorbent (ELISA) blocking assay. J Clin Microbiol 8:283-287, 1978c. Yolken, R. H., Stopa, P. J.: Enzyme-linked fluorescence assay: ultrasensitive solid-phase assay for detection of human rotavirus. J Clin Microbiol 10:317—321, 1979. Zissis, G., Lambert, J. P.: Enzyme—linked immunosorbent assays adapted for serotyping of human rotavirus strains. J Clin Microbiol 11:1—5, 1980. Zissis, G., Lambert, J. P., Marbehant, P., Marissens, D., Lobmann, M., Charlier, P., Delem, A., Zygraichi, N.: Protection studies in colostrum—deprived piglets of a bovine rotavirus vaccine candidate using human rotavirus strains for challenge. J Infect Dis 148:1061-1068, 1983. VITA DC at of Ve Mi: au' M311 Sc: In ass 197 136 VITA The author was born in St. Paul, Minnesota, on October 1, 1953. He completed his high school education at Columbia Heights, Minnesota. He received the Bachelor of Science degree in 1976 and the degree of Doctor of Veterinary Medicine in June, 1978, from the University of Minnesota. After one year of private veterinary practice, the author accepted a position as resident/instructor at Michigan State University. The residency and a Master of Science degree in pathology were completed in June, 1982. In July, 1982, the author began employment as an assistant professor in the Department of Veterinary Science at South Dakota State University. The author was married to Barbara Bjorke Collins in 1974 and they have two sons, Brian and Daniel. “mm111M)»mmmgagmm“