Next Article in Journal
Understanding Bacteriophage Specificity in Natural Microbial Communities
Previous Article in Journal
Th17 Lymphocytes in Respiratory Syncytial Virus Infection
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Mutation Distribution in the NSP4 Protein in Rotaviruses Isolated from Mexican Children with Moderate to Severe Gastroenteritis

Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, Av. Universidad S/N Ciudad Universitaria, San Nicolás de los Garza, Nuevo León, CP. 66451, Mexico
*
Author to whom correspondence should be addressed.
Viruses 2013, 5(3), 792-805; https://doi.org/10.3390/v5030792
Submission received: 13 February 2013 / Revised: 4 March 2013 / Accepted: 5 March 2013 / Published: 11 March 2013

Abstract

:
The NSP4 protein is a multifunctional protein that plays a role in the morphogenesis and pathogenesis of the rotavirus. Although NSP4 is considered an enterotoxin, the relationship between gastroenteritis severity and amino acid variations in NSP4 of the human rotavirus remains unclear. In this study, we analyzed the sequence diversity of NSP4 and the severity of gastroenteritis of children with moderate to severe gastroenteritis. The rotavirus-infected children were hospitalized before the rotavirus vaccine program in Mexico. All children had diarrhea within 1−4 days, 44 (88%) were vomiting and 35 (70%) had fevers. The severity analysis showed that 13 (26%) cases had mild gastroenteritis, 23 (46%) moderate gastroenteritis and 14 (28%) severe. NSP4 phylogenetic analysis showed three clusters within the genotype E1. Sequence analysis revealed similar mutations inside each cluster, and an uncommon variation in residue 144 was found in five of the Mexican NSP4 sequences. Most of the amino acid variations were located in the VP4 and VP6 binding site domains, with no relationship to different grades of gastroenteritis. This finding indicates that severe gastroenteritis caused by the rotavirus appears to be related to diverse viral or cellular factors instead of NSP4 activity as a unique pathogenic factor.

1. Introduction

Rotaviruses cause gastroenteritis in almost all mammals and some birds [1]. Group A, B and C rotaviruses are known to infect humans and animals; however, group A is responsible for gastroenteritis in children less than five years old [2]. The common symptoms of this disease are diarrhea, fever and vomiting [3]. Dehydration is a consequence of severe diarrhea that may cause infant death [2,3]. Statistics reveal that rotaviruses cause approximately 453,000 child deaths per year worldwide, with the highest mortality rates primarily in developing countries [4]. The rotavirus pathogenesis is related to the non-structural protein 4 (NSP4), which is a known enterotoxin [5]. NSP4 induces an intracellular calcium imbalance, resulting in membrane instability and loss of water; the same effect would be present by phospholipase C-mediated inositol 1,4,5-trisphosphate production when NSP4 interacts with non-infected cells [2,6,7,8].
NSP4 is a glycosylated protein of 175 amino acids and a molecular mass of 28 kDa in its mature form. This protein is characterized by three hydrophobic domains named H1 (residues 7−21), H2 (29−47) and H3 (67−85) and a coiled α-helical domain (95−137) [9]. The NSP4 amino-terminal region (1−44) is located in the lumen of the endoplasmic reticulum, whereas its carboxy-terminal region (45–175) is in the cytoplasm and interacts with different proteins including VP6 and VP4 during rotavirus morphogenesis [10,11,12]. NSP4 also interacts with some cellular proteins and extracellular matrix proteins [13,14,15].
On the other hand, the NSP4 sequence analysis has revealed at least 14 genotypes named E1−E14 (for Enterotoxin). The human rotavirus genotypes are E1 (Wa-like), E2 (Kun-like) and E3 (AU-1), previously known as genotypes B, A and C, respectively [16]. Information related to rotavirus infection and the role of NSP4 pathogenesis in humans has not been described in detail. Some reports indicate that changes in the sequence of NSP4, VP4 and VP7 are related with asymptomatic strains isolated from humans [17,18], but amino acids variation in NSP4 has not always been associated with asymptomatic infections [19,20,21]. In this study, we analyzed NSP4 of human rotavirus strains in Mexican children with different grades of gastroenteritis to determine the genotype, distribution and frequency of mutations in NSP4.

2. Results and Discussion

2.1. Rotavirus Positive Samples and Gastroenteritis Severity

A total of 123 diarrheic feces collected from October 2004 to March 2005 from hospitalized children with gastroenteritis in Monterrey, Nuevo Leon, México, were analyzed to detect rotavirus. Of all the analyzed samples, sixty-six (53.7%) were positive for the presence of a rotavirus. This is a high percentage, because usually rotavirus infection is associated with 25−39% of hospitalizations for acute gastroenteritis [22]. To further analyze the gastroenteritis severity, 16 of the 66 rotavirus positive samples were discarded due to incomplete data about the infected children or their symptoms. The remaining 50 gastroenteritis cases were considered in this study and the signs and symptoms of the rotavirus gastroenteritis were examined. Twenty-eight (56%) cases were male children and 22 (44%) were female children. Most of the rotavirus gastroenteritis cases (86%) were related to children under two years old, this is in agreement with previous reports on rotavirus infection [23,24,25]. All the infected children included in this study had diarrhea within 1 to 4 days, 44 (88%) of them were vomiting and 35 (70%) had a fever. The results of the rotavirus gastroenteritis severity showed that 13 (26%) cases had a score ≤ 10, whereas 23 (46%) infections showed a score ≥ 11 and 14 (28%) cases had a score ≥ 15. Those scores were grouped as mild, moderate and severe gastroenteritis, respectively (Table 1). Although some studies describe breastfeeding as an important factor to avoid severe gastroenteritis, we did not find a relationship between breastfeeding and the rotavirus gastroenteritis severity [26,27,28]. This type of analysis of rotavirus regarding the prevalence and severity of the gastroenteritis may represent a basis to compare the epidemic seasons of the rotavirus before and after the introduction of the Rotarix® vaccine (GSK) [29]. In this aspect, some studies have shown that this vaccine can diminish the cases of hospitalization by the rotavirus from 40 to 60% [30,31,32]. Additionally, studies in Latin America have shown the efficiency of the vaccine to decrease the severity of the rotavirus gastroenteritis [33,34].

2.2. The NSP4 Genotype

NSP4 is a main factor of rotavirus pathogenesis [35,36]. Therefore, in this study we have focused on the analysis of the NSP4 genotype and its sequence relationship with the rotavirus gastroenteritis severity. To amplify and identify the NSP4 genotype in all the rotavirus positive samples we used a combination of different primers that have been previously described [20,21,37]. This was a useful strategy that identified 61 (92.4%) of the samples as NSP4 genotype E1. The predominant NSP4 genotype E1 identified in the studied area is commonly reported; some studies identified the E2 genotype as the second common genotype [38,39,40,41]. Furthermore, others reported non-common NSP4 genotypes E3, E5, E6 and E13 in human rotavirus strains in Thailand, Brazil, Bangladesh and Kenya [42,43,44,45,46].

2.3. The NSP4 Sequence Analysis

According to the gastroenteritis severity analysis (Table 1), the rotavirus positive samples were classified as mild (26%), moderate (46%) and severe (28%). Based on these results a stratified random sampling was done to select representative samples to the NSP4 sequence analysis, this selection included 4(31%) of 13 samples of mild cases, 7 (30%) of 23 cases of moderate and 5 (36%) of 14 severe cases. The analysis of the deduced amino acid sequences of NSP4 reported in this study showed three clusters inside the same genotype E1 (Figure 1). The NSP4 sequences MX04-29, MX05-58, MX05-126 reported here grouped in the cluster I; the samples MX05-48, MX05-71, MX05-88, MX05-137, and MX05-144 were in the cluster II and the samples MX04-27, MX04-28, MX05-36, MX05-51, MX05-64, MX05-68, MX05-107 and MX05-119 in the cluster III (Table 3). Previous reports have shown the presence of at least two clusters within this genotype, and in some of them the clusters were related to the location or to the isolation date of the rotavirus strains, in this study we did not observed such relation [41,47,48]. The NSP4 amino acids variations showed in the cluster I (amino acids AK in position 136-137 respectively) were related with rotavirus strains reported in Italy, China, Spain, United States of America (USA) and Russia (Accession number ACF77154, AFU36983, ADU55685, ADO78536 and ACY01369). The sequences in the cluster II share the same amino acid variations in the positions 141-145 with sequences from China, Russia, Thailand and USA (AAOO6852, ACQ99541, AFQ20926, ADO78564) and the cluster III shared common amino acid variations in aa 141,142, 144 and 145 with the strain Vanderbilt isolated in USA (AEB80046). Further analyses on NSP4 were performed using the amino acid frequency in each specific position in the protein. The analysis of 349 NSP4 sequences from the GenBank database showed that this protein is highly conserved in some specific domains (Table 2). These conserved regions include the glycosylation sites (aa 8 and 18), the hydrophobic region H1, H2 and H3 (aa 7−21, 29−47 and 67−85), the transmembrane domain (aa 22−44) and the coiled α-helical domain (aa 97−137) where the frequency of the consensus amino acid in a specific position was 97.6 to 100%; however, the H3 domain showed an amino acid frequency of 88.9% for I72 and 87.4% for I76 ( Table 2). Conversely, punctual variations in the NSP4 sequence fell in the VP4 binding site domain where the lowest amino acid frequency was at position 141 with a valine present for 58% of the studied sequences, and also the VP6 binding site where a serine at position 169 had a frequency of 56% ( Table 2). Most of the amino acids variations in the NSP4 sequences reported were positioned in the carboxyl terminal region (Table 3). However, the samples MX04-29, MX05-58 and MX05-126 showed punctual variations in conserved amino acids 111, 136 and 137 (85.7−92.8%), respectively (Table 3). In addition, five of the NSP4 sequences reported in this study had an uncommon amino acid change at position 144, where a methionine was replaced by a valine. Usually methionine is present in this position in 97.9% of all the 349 NSP4 sequences analyzed (Figure 1), and thus this amino acid variation is unique in our sequences. Moreover, we did not find valine in this position in any other NSP4 genotype E1 sequence in the NCBI database. The replacement of a methionine may be not significant when it is replaced by another hydrophobic amino acid such as valine, because both amino acids can play a role in binding or recognition of hydrophobic ligands such as lipids. However, the sulfur atom in methionine can be involved in binding metals [49] and NSP4 presents a metal binding domain between residues 114 and 135 [50,51]; nevertheless, further analysis is required to explain the importance of the mutations in such conserved amino acid position within NSP4.
Table 1. Characteristics and symptoms of the children infected with the rotavirus and the analysis of the rotavirus severity score from slight to severe according to the Ruuska score [52].
Table 1. Characteristics and symptoms of the children infected with the rotavirus and the analysis of the rotavirus severity score from slight to severe according to the Ruuska score [52].
Gastroenteritis
severity
(Ruuska score)
Gastroenteritis severityIncidenceBreastfeedingSexAge*
(months)
diarrhea episodes* /24 hDays with diarrhea*Vomiting episodes* / 24hrsDays of vomiting*
MF
≤ 10Mild13 (26%)61.5%6786.12.42.61.0
≥ 11Moderate23 (46%)82.3%158128.13.14.82.7
≥ 15Severe14 (28%)71.4%771410.44.39.33.6
* Average Data
Table 2. Consensus NSP4 sequence and the frequency of each amino acid in a specific position from residues 1 to 175. The analysis was obtained from 349 NSP4 sequences from the GenBank and is complemented with the description of some of the NSP4 domains. Abbreviations: GS: Glycosylation site, H: Hydrophobic domain.
Table 2. Consensus NSP4 sequence and the frequency of each amino acid in a specific position from residues 1 to 175. The analysis was obtained from 349 NSP4 sequences from the GenBank and is complemented with the description of some of the NSP4 domains. Abbreviations: GS: Glycosylation site, H: Hydrophobic domain.
Transmembrane site (22−44)
GS1H1 (aa 7−21)GS2
Consensus seqMDKLADLNYTLSVITLMNDTLHSII
Frequency %10098.599.79897.699.510099.710099.499.492.799.797.998.29910099.497.998.810098.898.810098.8
aa position26272829303132333435363738394041424344454647484950
H2 (29−47)
Transmembrane domain (22−44)
Consensus seq.QDPGMAYFPYIASVLTVLFTLHKAS
Frequency %97.699.710010096.410010010097.810098.810099.799.410010099499.499.798.410099.499.410099.4
aa position51525354555657585960616263646566676869707172737475
H3 ( aa 67-85)
Consensus seq.IPTMKIALKTSKCSYKVIKYCIVTI
Frequency %99.499.499.19999.799.410099.496.699.410098.899.110099.710099.497.999.198.598.888.999.199.495.5
aa position767778798081828384858687888990919293949596979899100
H3 (67−85)Alpha coiled Domain (95−137)
Consensus seq.INTLLKLAGYKEQVTTKDEIEQQMD
Frequency %87.499.798.510099.198.898.298.810099.498.510099.196.798.89899.198.598.899.710098.299.799.799.1
aa position101102103104105106107108109110111112113114115116117118119120121122123124125
Alpha coiled Domain (95−137)
VP4 Binding site (112−148)
Enterotoxin (114-135)
Consensus seq.RIVKEMRRQLEMIDKLTTREIEQVE
Frequency %99.798.899.19999.799.499.710098.599.793.499.499.799.199.110010098.899.199.799.799.799.799.4100
aa position126127128129130131132133134135136137138139140141142143144145146147148149150
Alpha coiled Domain (95−137)
VP4 Binding site (112−148)
Enterotoxin (114−135)
Consensus seq.LLKRIHDNLITRPVDVIDMSKEFNQ
Frequency %99.410099.79999.792.899.178.610090.793.487.178.48889.8587699.497.964.399.199.197.399.1100
aa position151152153154155156157158159160161162163164165166167168169170171172173174175
VP6 Binding site (167−175)
Consensus seq.KNIKTLDEWESGKNPYEPSEVTASM
Frequency %98.899.7949310099.798.299.110095.87799.797.699.110010099.11005699.199.499.710098.8100
Table 3. Sequence analysis of the amino acid variations in the NSP4 protein from rotavirus strains reported in this study.
Table 3. Sequence analysis of the amino acid variations in the NSP4 protein from rotavirus strains reported in this study.
NSP4 Sequence Severity scores ClusterAmino acid variability and distribution
H3 *TDACD *E/VP4 *VP4 *VP6 *
76778594111132136137141142144145161169172173174175
MX04-2916I N AK
MX05-5814I D AK
MX05-12616I V D AK
MX05-488IIV H T TNI
MX05-14413IIV T TNI
MX05-13715IIV T TNI
MX05-8816IIV T TNI
MX05-710IIV T TNM
MX04-2711III SVVT I
MX04-2814III SVVT I
MX05-368III SVVT I
MX05-5114III N SVVT II
MX05-6412III SVVT I
MX05-688III G SVVT I
MX05-10714III SVVT I
MX05-11915III SVVTNI
Consensus amino acids I N Y E E D T R V I M S S S T A S M
Frequency % 86 99.7 99.4 99.1 89.4 99.4 92.8 85.7 56.2 75.9 99.4 61 76 56 100 100 99 100
* H3: hydrophobic region 3; TD: Tetramerization Domain; ACD: Alpha Coiled Coil Domain E: Enterotoxin Domain; VP4: VP4 Binding site domain; VP6: VP6 binding site domain
Figure 1. Phylogenetic analysis of the deduced amino acid NSP4 sequences reported in this study and other NSP4 genotypes previously reported in the NCBI. The phylogenetic tree was constructed based on the neighbor-joining method. The bootstrap consensus tree inferred from 500 replicates is taken to represent the evolutionary history of the taxa analyzed. The evolutionary distances were computed using the p-distance method and are in the units of the number of amino acid differences per site. Evolutionary analyses were conducted in MEGA5 package [54]. Accession number of the sequences reported in this study in the GenBank: MX04-28: JX458969, MX04-29: JX458970, MX04-27:JX458971, MX05-51:JX458972, MX05-144: JX458973, MX05-58: JX458974, MX05-68: JX458975, MX05-71:JX458976, MX05-88:JX458977, MX05-119:JX458978, MX05-107: JX458982, MX05-126: JX458983, MX05-137: JX458984.
Figure 1. Phylogenetic analysis of the deduced amino acid NSP4 sequences reported in this study and other NSP4 genotypes previously reported in the NCBI. The phylogenetic tree was constructed based on the neighbor-joining method. The bootstrap consensus tree inferred from 500 replicates is taken to represent the evolutionary history of the taxa analyzed. The evolutionary distances were computed using the p-distance method and are in the units of the number of amino acid differences per site. Evolutionary analyses were conducted in MEGA5 package [54]. Accession number of the sequences reported in this study in the GenBank: MX04-28: JX458969, MX04-29: JX458970, MX04-27:JX458971, MX05-51:JX458972, MX05-144: JX458973, MX05-58: JX458974, MX05-68: JX458975, MX05-71:JX458976, MX05-88:JX458977, MX05-119:JX458978, MX05-107: JX458982, MX05-126: JX458983, MX05-137: JX458984.
Viruses 05 00792 g001

3. Experimental Section

3.1. Samples Recollection and Gastroenteritis Severity Score

A total of 123 stool samples were collected from hospitalized children with gastroenteritis in Monterrey, Nuevo León, México, from October 2004 to March 2005. The inclusion criteria were the age of the child, up to five years old, and the hospitalization for nonbacterial gastroenteritis. Diarrhea, vomiting and fever were used as registered symptoms to calculate the gastroenteritis severity, and formed the basis of the Ruuska score [52].

3.2. Extraction of Rotavirus RNA.

The feces samples were used to isolate, purify and detect the rotavirus RNA genome by TRI Reagent® (Molecular Research Center, Cincinnati, OH) as suggested the by supplier. The viral RNA was loaded onto a 10% polyacrylamide gel under native conditions, and then stained by a silver-staining procedure. A sample was considered positive to the rotavirus when a characteristic double-stranded RNA genome was observed in the gel [53].

3.3. NSP4 Genotype Identification and Sequence

RNA positive samples for the rotavirus were retro transcribed and amplified by PCR to isolate the NSP4 gene using the primers Beg16-End722 or NSP41F-NSP42R [20,21]; although in several experiments, combinations of both primer pairs were required to achieve amplification. NSP4 genotype identification was performed by a multiplex-seminested PCR, with 10END722 or NSP42R as the external primers and Wa, Kun or RRV as the internal primers, which corresponds to genotypes E1, E2 and E3, respectively [37]. Samples of the amplified NSP4 gene were cloned using the pGEM-T vector (Promega Inc, Madison, WI) according to the manufacturer´s instructions. The plasmid with the NSP4 insert was purified by the Wizard SV Minipreps kit (Promega Inc, Madison, WI) and sequenced by the dideoxynucleotide chain termination method, using an ABI Prism Big Dye Terminator Cycle Sequencing Ready Reaction kit (PE Applied Biosystems, Whashington, DC). The DNA sequence was confirmed by sequencing both DNA strands of each of the different clones using the pUCM13 sense and antisense standard primers. The resulting sequences were analyzed with MEGA 5.0 and compared with other sequences reported in the GenBank data base; the phylogenetic tree was determined by the Neighbor joining method [54]. The GenBank accession numbers of NSP4 sequences used in sequence analysis were AB361285, AB008233, AB008237, AB213391, AB326290, AB326294, AB326963, AB361282, AF170830, AF260930, AY159640, AY159642, AY353740, AY353800, DQ909069, DQ909070, EF033202, EF033203, EF672575, EU679377, EU679382, U42628, U83798, AAO06852, AAT48079, AB008229 - AB008231, AB008234-AB008236, AB008238-AB008245, AB008247-AB008257, AB008259 - AB008263, AB022772, AB043026, AB043069-AB043078, AB196491, AB196492, AB196958, AB196959, AB211987-AB213392, AB232699, AB269688, AB303218, AB326286-AB326289, AB326293, AB326295, AB326297, AB326334, AB326336, AB326337, AB326347, AB326348, AB326962, AB326966, AB326969, AB326971, AB361276, AB361281, AB361284, AB361286-AB361288, ABK62862, ABU49806, ACF77153, ACF77154, ACJ54826, ACJ66758, ACJ66769, ACL80635, ACL80638, ACQ99541, ACY01369, ACY01381, ACZ51671, ADA70484, ADK46705, ADK46715, ADO78533, ADO78536, ADO78564, AEB79485, AEB79550, AEK69633, AET43468, AF161810-AF161815, AF170831-AF170833, AF173179, AF173181-AF173208, AF173211 - AF173214, AF174300 - AF174302, AF260928, AF260929, AF284776-AF284778, AF469676 - AF469679, AF506016, AF541921, AFJ68184, AFJ68397, AFK27432, AJ236757 - AJ236770, AJ236772 - AJ236774, AJ236778 - AJ236782, AJ400634, AY159630 - AY159632, AY159634 - AY159639, AY159641, AY159643, AY159644 - AY159647, AY353727, AY353728, AY353730 - AY353739, AY353741 - AY353746, AY353753 - AY353765, AY353767 - AY353790, AY353792 - AY353805, AY601540 - AY601544, AY629562, BAD84188, BAF97950, CAB36938, D88830, DQ146647, DQ146658, DQ146669, DQ146680, DQ189233 - DQ189237, DQ189240, DQ299876, DQ339147 - DQ339151, DQ490543, DQ492678, DQ525182 - DQ525188, EF011980, EF033204, EF059918, EF059919, EF059924, EF159574, EU679378 - EU679380, GAU78558, Q9YJN7, U59108, U59110.

4. Conclusions

The presence of intra-genotypic clusters and punctual amino acid variations in the NSP4 genotype E1 may indicate that NSP4 mutates mainly via accumulation of single point mutations. Since most of the variations in NSP4 fell in the carboxylic terminal region, especially in the VP4 binding site segment, it is important to consider that NSP4 is involved in morphogenesis and pathogenesis activities. Further analysis of NSP4 in the VP4 and VP6 binding site segment should be studied, especially with respect to structural conformational changes caused by amino acid variations. NSP4 is an important factor in rotavirus pathogenesis, and in this study an analysis examining the amino acid variations in the sequence and the gastroenteritis severity score was performed. The results failed to show a relationship between punctual variations in NSP4 and the severity of rotavirus gastroenteritis. The study of the NSP4 protein and its interaction with other viral proteins may aid our understanding of the pathogenesis of the rotavirus.

Conflict of Interest

The authors declare no conflict of interest

References and Notes

  1. Kapikian, A.; Hoshino, Y.; Chanock, R. Rotaviruses. In Fields virology, 4th; Knipe, D.M., Howley, P.M., Griffin, D.E., Lamb, R.A., Martin, M.A., Roizman, B., Strais, S.E., Eds.; Lippincott Williams and Wilkins: Philadelphia, Pennsylvania Pa, USA, 2001; pp. 1787–1833. [Google Scholar]
  2. Morris, A.P.; Estes, M.K. Microbes and microbial toxins: Paradigms for microbial mucosal interactions VIII: Pathological consequences of rotavirus infection an enterotoxin. Am. J. Physiol.-Gastr. L. 2001, 281, 303–310. [Google Scholar]
  3. Ramig, R.F. Pathogenesis of intestinal and systemic rotavirus infection. J. Virol. 2004, 78, 10213–10220. [Google Scholar] [CrossRef]
  4. Tate, J.E.; Burton, A.H.; Boschi-Pinto, C.; Steele, A.D.; Duque, J.; Parashar, U.D. 2008 estimate of worldwide rotavirus-associated mortality in children younger than 5 years before the introduction of universal rotavirus vaccination programmes: A systematic review and meta-analysis. Lancet Infect. Dis. 2012, 12, 131–141. [Google Scholar]
  5. Ball, J.M.; Tian, P.; Zeng, C.Q.; Morris, A.P.; Estes, M.K. Age-dependent diarrhea induced by a rotaviral nonstructural glycoprotein. Science 1996, 272, 101–104. [Google Scholar]
  6. Dong, Y.; Zeng, C.; Ball, J.; Estes, M.; Morris, A. The rotavirus enterotoxin NSP4 mobilizes intracellular calcium in human intestinal cells by stimulating phospholipase C-mediated inositol 1,4,5-trisphosphate production. Proc. Natl. Acad. Sci. USA 1997, 94, 3960–3965. [Google Scholar] [CrossRef]
  7. Brunet, J.P.; Cotte, J.; Linxe, C.; Quero, A.M; Géniteau-Legendre, M.; Servin, A. Rotavirus infection induces an increase in intracellular calcium concentration in human intestinal epithelial cells: Role in microvillar actin alteration. J. Virol. 2000, 74, 2323–2332. [Google Scholar]
  8. Zhang, M.; Zeng, C.Q.; Morris, A.P.; Estes, M.K. A functional NSP4 enterotoxin peptide secreted from rotavirus-infected cells. J. Virol. 2000, 74, 663–11670. [Google Scholar]
  9. Estes, M.K. Rotaviruses and their replication. In Fields virology, 4th; Knipe, D.M., Howley, P.M., Griffin, D.E., Lamb, R.A., Martin, M.A., Roizman, B., Strais, S.E., Eds.; Lippincott Williams and Wilkins: Philadelphia, Pennsylvania, USA, 2001; pp. 1747–1785. [Google Scholar]
  10. Au, K.; Chan, W.; Burns, J.W.; Estes, M.K. Receptor activity of rotavirus nonstructural glycoprotein NS28. J. Virol. 1989, 63, 4553–4562. [Google Scholar]
  11. Bergmann, C.C.; Maas, D.; Poruchynsky, M.S.; Atkinson, P.H.; Bellamy, A.R. Topology of the non-structural rotavirus receptor glycoprotein NS28 in the rough endoplasmic reticulum. EMBO J. 1989, 8, 1695–1703. [Google Scholar]
  12. Tian, P.; Ball, J.; Zeng, C.; Estes, M. Rotavirus Protein Expression is important for virus assembly and pathogenesis. Arch. Virol. 1996, 12, 69–77. [Google Scholar]
  13. Mirazimi, A.; Nilsson, M.; Svensson, L. The molecular chaperone calnexin interacts with the NSP4 enterotoxin of rotavirus in vivo and in vitro. J. Virol. 1998, 72, 8705–8709. [Google Scholar]
  14. Xu, A.; Bellamy, A.R.; Taylor, J.A. Immobilization of the early secretory pathway by a virus glycoprotein that binds to microtubules. EMBO J. 2000, 19, 6465–6474. [Google Scholar] [CrossRef]
  15. Storey, S.M.; Gibbons, T.F.; Williams, C.V.; Parr, R.D.; Schroeder, F.; Ball, J.M. Full-length, glycosylated NSP4 is localized to plasma membrane caveolae by a novel raft isolation technique. J. Virol. 2007, 81, 5472–5483. [Google Scholar]
  16. Matthijnssens, J.; Ciarlet, M.; McDonald, S.M.; Attoui, H.; Bányai, K.; Brister, J.R.; Buesa, J.; Esona, M.D.; Estes, M.K.; Gentsch, J.R.; et al. Uniformity of rotavirus strain nomenclature proposed by the Rotavirus Classification Working Group (RCWG). Arch. Virol. 2011, 156, 1397–1413. [Google Scholar] [CrossRef]
  17. Kirkwood, C.D.; Coulson, B.; Bishop, R. G3P2 rotaviruses causing diarrhoeal disease in neonates differ in VP4, VP7 y NSP4 sequence from G3P2 strains causing asymptomatic neonatal infection. Arch. Virol. 1996, 141, 1661–1676. [Google Scholar] [CrossRef]
  18. Pager, C.T.; Alexander, J.J.; Steele, A.D. South African G4P[6] asymptomatic and symptomatic neonatal rotavirus strains differ in their NSP4, VP8*, and VP7 genes. J. Med. Virol. 2000, 62, 208–216. [Google Scholar] [CrossRef]
  19. Horie, Y.; Masamune, O.; Nakagomi, O. Three major alleles of rotavirus NSP4 proteins identified by sequence analysis. J. Gen. Virol. 1997, 78, 2341–2346. [Google Scholar]
  20. Ward, R.L.; Mason, B.B.; Bernstein, D.J.; Sander, D.S.; Smith, V.E.; Zandle, G.A.; Rappaport, R.S. Attenuation of a human rotavirus vaccine candidate did not correlate with mutations in the NSP4 protein gene. J. Virol. 1997, 71, 6267–6270. [Google Scholar]
  21. Lee, C.; Wang, Y.; Kao, C.; Zao, C.; Lee, C.; Chen, H. NSP4 gene analysis of rotaviruses recovered from infected children with and without diarrhea. J. Clin. Microbiol. 2000, 38, 4471–4477. [Google Scholar]
  22. Parashar, U.D.; Gibson, C.J.; Bresee, J.S.; Glass, R.I. Rotavirus and severe childhood diarrhea. Emerg. Infect. Dis. 2006, 12, 304–306. [Google Scholar] [CrossRef]
  23. Linhares, A.C.; Velázquez, F.R.; Pérez-Schael, I.; Sáez-Llorens, X.; Abate, H.; Espinoza, F.; López, P.; Macías-Parra, M.; Ortega-Barría, E.; Rivera-Medina, D.M.; et al. Efficacy and safety of an oral live attenuated human rotavirus vaccine against rotavirus gastroenteritis during the first 2 years of life in Latin American infants: Randomised, double-blind controlled study. Lancet 2008, 371, 1181–1189. [Google Scholar] [CrossRef]
  24. Forster, J.; Guarino, A.; Parez, N.; Moraga, F.; Román, E.; Mory, O.; Tozzi, A.E.; López de Aguileta, A.; Wahn, U.; Graham, C.; et al. Hospital-Based Surveillance to Estimate the Burden of Rotavirus Gastroenteritis Among European Children Younger Than 5 Years of Age. Pediatrics 2009, 123, 393–400. [Google Scholar] [CrossRef]
  25. Bruijning-Verhagen, P.; Quach, C.; Bonten, M. Nosocomial Rotavirus Infections: A Meta-analysis. Pediatrics 2012, 129, 1011–1019. [Google Scholar] [CrossRef]
  26. Clemens, J.; Rao, M.; Ahmed, F.; Ward, R.; Huda, S.; Chakraborty, J.; Yunus, M.; Khan, M.R.; Ali, M.; Kay, B.; et al. Breastfeeding and the risk of life-threatening rotavirus diarrhea: Prevention or postponement? Pediatrics 1993, 92, 680–685. [Google Scholar]
  27. Molyneaux, P. Human immunity to rotavirus. J. Med. Microbiol. 1993, 43, 397–404. [Google Scholar] [CrossRef]
  28. Raisler, J.; Alexander, C.; O'Campo, P. Breast-feeding and infant illness: A dose-response relationship? Am. J. Public Health 1999, 89, 25–30. [Google Scholar] [CrossRef]
  29. Ward, R.L.; Bernstein, D.I. Rotarix: A rotavirus vaccine for the world. Clin. Infect. Dis. 2009, 48, 222–228. [Google Scholar] [CrossRef]
  30. De Palma, O.; Cruz, L.; Ramos, H.; de Baires, A.; Villatoro, N.; Pastor, D.; de Oliveira, L.H.; Kerin, T.; Bowen, M.; Gentsch, J.; et al. Effectiveness of rotavirus vaccination against childhood diarrhoea in El Salvador: Case-control study. BMJ 2010, 340, 2825–2832. [Google Scholar] [CrossRef]
  31. Munos, M.K.; Fischer, C.L.; Black, R.E. The effect of rotavirus vaccine on diarrhoea mortality. Int. J. Epidemiol. 2010, 39, 56–62. [Google Scholar] [CrossRef]
  32. Anderson, E.J.; Rupp, A.; Shulman, S.T.; Wang, D.; Zheng, X.; Noskin, G.A. Impact of rotavirus vaccination on hospital-acquired rotavirus gastroenteritis. Pediatrics 2011, 127, 264–270. [Google Scholar] [CrossRef]
  33. Salinas, B.; Pérez-Schael, I.; Linhares, A.C.; Ruiz-Palacios, G.M.; Guerrero, M.L.; Yarzábal, J.P.; Cervantes, Y.; Costa-Clemens, S.; Damaso, S.; Hardt, K.; De Vos, B. Evaluation of safety, immunogenicity and efficacy of an attenuated rotavirus vaccine, RIX4414: A randomized, placebo-controlled trial in Latin American infants. Pediatr. Infect. Dis. J. 2005, 24, 807–16. [Google Scholar] [CrossRef]
  34. Ruiz-Palacios, G.M.; Perez-Schael, I.; Velazquez, F.R.; Abate, H.; Breuer, T.; Costa-Clemens, S.; Cheuvart, B.; Espinoza, F.; Gillard, P.; Innis, B.L.; et al. Safety and efficacy of an attenuated vaccine against severe rotavirus gastroenteritis. N. Engl. J. Med. 2006, 354, 11–22. [Google Scholar] [CrossRef]
  35. Ball, J.M.; Mitchell, D.M.; Gibbons, T.F.; Parr, R. Rotavirus NSP4: A Multifunctional Viral Enterotoxin. Viral Immunol. 2005, 18, 27–40. [Google Scholar] [CrossRef]
  36. Lorrot, M.; Vasseur, M. How do the rotavirus NSP4 and bacterial enterotoxins lead differently to diarrhea? Virol. J. 2007, 4, 31–37. [Google Scholar] [CrossRef]
  37. Kudo, S.; Zhou, Y.; Cao, X.; Yamanishi, S.; Nakata, S.; Ushijima, H. Molecular characterization in the VP7, VP4 and NSP4 genes of human rotavirus serotype 4 (G4) isolated in Japan and Kenya. Microbiol. Immunol. 2001, 45, 167–171. [Google Scholar]
  38. Tavares, T.M.; Brito, W.M.E.D.; Fiaccadori, F.S.; Leal de Freitas, E.R.; Parente, J.A.; Sucasas da Costa, P.S.; Giugliano, L.G.; Andreasi, M.S.A.; Soares, C.M.A.; Cardoso, D.D.P. Molecular characterization of the NSP4 gene of human group A rotavirus samples from the West Central region of Brazil. Mem. Inst. Oswaldo Cruz 2008, 103, 288–294. [Google Scholar] [CrossRef]
  39. Bányai, K.; Bogdán, A.; Szucs, G.; Arista, S.; De Grazia, S.; Kang, G.; Banerjee, I.; Iturriza-Gómara, M.; Buonavoglia, C.; Martella, V. Assignment of the group A rotavirus NSP4 gene into genotypes using a hemi-nested multiplex PCR assay: A rapid and reproducible assay for strain surveillance studies. J. Med. Microbiol. 2009, 58, 303–311. [Google Scholar] [CrossRef] [Green Version]
  40. Vizzi, E.; Piñeros, O.; González, G.G.; Zambrano, J.L.; Lurdet, J.E.; Liprandi, F. Genotyping of human rotaviruses circulating among children with diarrhea in Valencia, Venezuela. J. Med. Virol. 2011, 83, 2225–2232. [Google Scholar] [CrossRef]
  41. Ben Hadj Fredj, M.; Zeller, M.; Fodha, I.; Heylen, E.; Chouikha, A.; Van Ranst, M.; Matthijnssens, J.; Trabelsi, A. Molecular characterization of the NSP4 gene of human group A rotavirus strains circulating in Tunisia from 2006 to 2008. Infect. Genet. Evol. 2012, 12, 997–1004. [Google Scholar] [CrossRef]
  42. Rahman, M.; Matthijnssens, J.; Yang, X.; Delbeke, T.; Arijs, I.; Taniguchi, K.; Iturriza-Gómara, M.; Iftekharuddin, N.; Azim, T.; Van Ranst, M. Evolutionary history and global spread of the emerging G12 human rotaviruses. J. Virol. 2007, 81, 2382–2390. [Google Scholar] [CrossRef]
  43. Matthijnssens, J.; Ciarlet, M.; Heiman, E.; Arijs, I.; Delbeke, T.; McDonald, S.M.; Palombo, E.A.; Iturriza-Gómara, M.; Maes, P.; Patton, J.T.; et al. Full genome-based classification of rotaviruses reveals a common origin between human Wa-Like and porcine rotavirus strains and human DS-1-like and bovine rotavirus strains. J. Virol. 2008, 82, 3204–3219. [Google Scholar] [CrossRef]
  44. Benati, F.J.; Maranhao, A.G.; Lima, R.S.; da Silva, RC.; Santos, N. Multiple-gene characterization of rotavirus strains: Evidence of genetic linkage among the VP7-, VP4-, VP6-, and NSP4-encoding genes. J. Med. Virol. 2010, 82, 1797–1802. [Google Scholar] [CrossRef]
  45. Khamrin, P.; Maneekarn, N.; Malasao, R.; Nguyen, T.A.; Ishida, S.; Okitsu, S.; Ushijima, H. Genotypic linkages of VP4, VP6, VP7, NSP4, NSP5 genes of rotaviruses circulating among children with acute gastroenteritis in Thail. Infect. Genet. Evol. 2010, 10, 467–472. [Google Scholar]
  46. Ghosh, S.; Gatheru, Z.; Nyangao, J.; Adachi, N.; Urushibara, N.; Kobayashi, N. Full genomic analysis of a simian SA11-like G3P[2] rotavirus strain isolated from an asymptomatic infant: Identification of novel VP1, VP6 and NSP4 genotypes. Infect. Genet. Evol. 2011, 11, 57–63. [Google Scholar] [CrossRef]
  47. Kirkwood, C.D.; Gentsch, J.R.; Glass, R.I. Sequence analysis of the NSP4 gene from human rotavirus strains isolated in the United States. Virus Genes 1999, 19, 113–122. [Google Scholar] [CrossRef]
  48. Araujo, I.T.; Heinemann, M.B.; Mascarenhas, J.D.P.; Santos Assis, R.M.; Fialho, A.M.; Leite, J.P.G. Molecular analysis of the NSP4 and VP6 genes of rotavirus strains recovered from hospitalized children in Rio de Janeiro, Brazil. J. Med. Microbiol. 2007, 56, 854–859. [Google Scholar] [CrossRef]
  49. Betts, M.J.; Russell, R.B. Amino Acid properties and Consequences of Substitutions. In Bioinformatics for Geneticists; Barnes, M.R., Gray, I.C., Eds.; Wiley: New York, New York, USA, 2003. [Google Scholar]
  50. Bowman, G.D.; Nodelman, I.M.; Levy, O.; Lin, S.L.; Tian, P.; Zamb, T.J.; Udem, S.A.; Venkataraghavan, B.; Schutt, C.E. Crystal structure of the oligomerization domain of NSP4 from rotavirus reveals a core metal-binding site. J. Mol. Biol. 2000, 304, 861–871. [Google Scholar] [CrossRef]
  51. Seo, N.; Zeng, CQ-Y, Hyser; Utama, B.; Crawford, S.E.; Kim, K.J.; Höök, M.; Estes, M.K. Integrins α1β1 and α2β1 are receptors for the rotavirus enterotoxin. Proc. Natl. Acad. Sci. USA 2008, 105, 8811–8818. [Google Scholar]
  52. Ruuska, T.; Vesikari, T. Rotavirus disease in Finnish children: use of numerical scoresfor clinical severity of diarrhoeal episodes. Scand. J. Infect. Dis. 1991, 22, 259–267. [Google Scholar] [CrossRef]
  53. Verly, E.; Cohen, J. Demostration of size variation of RNA segments between different isolates of calf rotavirus. J. Gen. Virol. 1977, 35, 583–586. [Google Scholar] [CrossRef]
  54. Tamura, K.; Peterson, D.; Peterson, N.; Stecher, G.; Nei, M.; Kumar, S. MEGA5: Molecular evolutionary genetics analysis using maximum likelihood, evolutionary distance, and maximum parsimony methods. Mol. Biol. Evol. 2011, 28, 2731–2739. [Google Scholar] [CrossRef]

Share and Cite

MDPI and ACS Style

González-Ochoa, G.; Menchaca, G.E.; Hernández, C.E.; Rodríguez, C.; Tamez, R.S.; Contreras, J.F. Mutation Distribution in the NSP4 Protein in Rotaviruses Isolated from Mexican Children with Moderate to Severe Gastroenteritis. Viruses 2013, 5, 792-805. https://doi.org/10.3390/v5030792

AMA Style

González-Ochoa G, Menchaca GE, Hernández CE, Rodríguez C, Tamez RS, Contreras JF. Mutation Distribution in the NSP4 Protein in Rotaviruses Isolated from Mexican Children with Moderate to Severe Gastroenteritis. Viruses. 2013; 5(3):792-805. https://doi.org/10.3390/v5030792

Chicago/Turabian Style

González-Ochoa, Guadalupe, Griselda E. Menchaca, Carlos E. Hernández, Cristina Rodríguez, Reyes S. Tamez, and Juan F. Contreras. 2013. "Mutation Distribution in the NSP4 Protein in Rotaviruses Isolated from Mexican Children with Moderate to Severe Gastroenteritis" Viruses 5, no. 3: 792-805. https://doi.org/10.3390/v5030792

Article Metrics

Back to TopTop