Skip to main content
Erschienen in: Virology Journal 1/2010

Open Access 01.12.2010 | Short report

A novel adenovirus of Western lowland gorillas (Gorilla gorilla gorilla)

verfasst von: Diana Wevers, Fabian H Leendertz, Nelly Scuda, Christophe Boesch, Martha M Robbins, Josephine Head, Carsten Ludwig, Joachim Kühn, Bernhard Ehlers

Erschienen in: Virology Journal | Ausgabe 1/2010

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Adenoviruses (AdV) broadly infect vertebrate hosts including a variety of primates. We identified a novel AdV in the feces of captive gorillas by isolation in cell culture, electron microscopy and PCR. From the supernatants of infected cultures we amplified DNA polymerase (DPOL), preterminal protein (pTP) and hexon gene sequences with generic pan primate AdV PCR assays. The sequences in-between were amplified by long-distance PCRs of 2 - 10 kb length, resulting in a final sequence of 15.6 kb. Phylogenetic analysis placed the novel gorilla AdV into a cluster of primate AdVs belonging to the species Human adenovirus B (HAdV-B). Depending on the analyzed gene, its position within the cluster was variable. To further elucidate its origin, feces samples of wild gorillas were analyzed. AdV hexon sequences were detected which are indicative for three distinct and novel gorilla HAdV-B viruses, among them a virus nearly identical to the novel AdV isolated from captive gorillas. This shows that the discovered virus is a member of a group of HAdV-B viruses that naturally infect gorillas. The mixed phylogenetic clusters of gorilla, chimpanzee, bonobo and human AdVs within the HAdV-B species indicate that host switches may have been a component of the evolution of human and non-human primate HAdV-B viruses.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1743-422X-7-303) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

DW and JK performed the cell culture experiments. DW, NS, JK and JH performed the molecular genetic studies. FHL, CB and MMR coordinated field work and sample collection. JH and CL sampled gorilla feces. DW, NS, JK, FHL and BE conceived of the study, and participated in its design and coordination. DW, FHL, CB, JK and BE drafted the manuscript. All authors read and approved the final manuscript.

Findings

Adenoviruses are non-enveloped icosahedral double-stranded DNA viruses that infect fish, amphibians, reptiles, birds and mammals [1]. Human adenoviruses (HAdV) are categorized into seven species (HAdV-A to HAdV-G) [2]. Each species includes a distinct number of serotypes [3]. In addition, intra-species shuffling of penton base, fiber and hexon genes by recombination has been frequently observed [46]. Simian adenoviruses have been discovered in monkeys and great apes [711]. They are very similar to HAdV, and most of them can be grouped into corresponding HAdV species or the newly established species Simian adenovirus A (SAdV-A).
In 2008, a group of Western lowland gorillas (Gorilla gorilla gorilla) suffered from prolonged diarrhea and self-limiting respiratory disease in the Zoological gardens of Münster, Germany. To isolate viral agents potentially responsible for the symptoms, fecal samples were suspended in phosphate-buffered saline, sterile filtered and cultured on MRC-5 cells and A549 cells. After eight days of culture, a cytopathogenic effect was observed. The culture supernatant was examined by electron microscopy, and virus-like structures were detected their size and general structure being consistent with that of adenoviruses (Additional Figure 1).
DNA was then extracted from culture supernatant using the Qiagen tissue kit according to the manufacturer's instructions (Qiagen, Hilden, Germany), and a generic primate adenovirus PCR was performed. For this purpose, a nested set of degenerate and deoxyinosine-substituted (deg/dI) primers was designed, targeting a highly conserved region of the DNA polymerase (DPOL) gene of primate mastadenoviruses (Figure 1; Table 1). PCR was performed in a total volume of 25 μl with 0.2 μl AmpliTaq Gold (Applied Biosystems), 20 pmol of each primer, 200 μM dNTPs, 2 mM MgCl2, and 5% DMSO. A T-Gradient thermocycler from Biometra was used with the following cycling conditions: 95°C for 12 min, and 45 cycles of 95°C for 30 sec, 45°C (1st round and 2nd round) for 30 sec and 72°C for 2 min, followed by a 15 min final extension step at 72°C. PCR products were purified using the Invisorb DNA clean up kit according to the instructions of the manufacturer (Invitek, Berlin, Germany), and directly sequenced with the Big Dye terminator cycle sequencing kit (Applied Biosystems, Warrington, UK) on a 377 automated DNA sequencer (Applied Biosystems). In BLAST analysis of GenBank, the sequence was most closely related to chimpanzee AdVs and human AdVs of the species HAdV-B. There was less similarity to the six gorilla HAdV-B viruses (SAdV-27.2; SAdV-28.2; SAdV-41.1; SAdV-41.2; SAdV-46; SAdV-47; Table 2) recently described [12]. Since the novel gorilla AdV was the seventh HAdV-B of this host, it was named for the purpose of this paper Gorilla gorilla adenovirus B7 (GgorAdV-B7).
Table 1
Primers for amplification of DPOL, pTP and hexon gene sequences
Primer-set abbreviation
Targeted gene
Name of primer
sequence 5'-3'
PCR length
Annealing temp.
Degenerate primers
DPOL-cons
DPOL
4431-s $
GTnTwyGAyAThTGyGGhATGTAyGC
  
  
4428-as
GAGGCTGTCCGTrTC(n/i)CCGTA#
956 bp
45°C
  
4428-s
CGGACGCCTCTGyTGGAC(n/i)AA
  
  
4429-as
GGCCAGCACrAA(n/i)GArGC
650 bp
45°C
Hex-5'-cons
Hexon
4515-s
GTGGATGG(n/i)GA(r/i)GG(n/i)TACA
  
  
4515-as
CGCACAACGTC(r/i)AA(n/i)AC(y/i)TC
536 bp
45°C
  
4516-s
TGTAACATGAC(y/i)AA(r(i)GA(y/i)TGG
  
  
4516-as
CAGGGCCCCCAT(n/i)GACA
381 bp
45°C
Hex3'-cons
Hexon
4517-s
CGCAATGGTC(n/i)TACATGCAC
  
  
4517-as
CAGTGCCCGA(r/i)TA(k/i)GG(n/i)TT
340 bp
45°C
  
4518-s
GCAGGACGC(y/i)TCGGAGTA
  
  
4518-as
CACCC(k/i)GTT(r/i)TC(n/i)CC
230 bp
45°C
pTP-cons
pTP
4521-s
TGGCGACGT(n/i)GT(n/i)TACAG
  
  
4521-as
CGGACT(y/i)(k/i)GA(r/i)CCTGAAA
260 bp
45°C
  
4522-s
TACAGCCG(n/i)GTSTGGAAC
  
  
4522-as
CTGAAAGAGAGTTC(n/i)ACAGAATCA
230 bp
45°C
Specific primers for long-distance PCR
pTP-DPOL-LD
pTP, DPOL
4659-s
TCGCATCTCCAACGACCT
  
  
4659-as
GCATCCATGGTGAAGATTCC
2350 bp
60°C
Hex-LD
Hexon
4618-s
AGTTCGCTACACACTGGCTG
  
  
4618-as
ATTGCGGTGATGATTGAATG
1354 bp
59°C
pTP-Hex-LD
pTP, Hexon
4662-s
CTCGGTATCGTTGACGGC
  
  
4662-as
GATCAACGGGCACAAAGC
10044 bp
60°C
Primers specific for AdV species B
Hex-loop2
Hexon
5442s
GAACAAGATACTTTAGCATGTGGAA
  
  
5442as
GATTGAATGGATTAACATTGTCC
468 bp
55°C
 
Hexon
5443s
TAGAAAATCACGGGGTGGAAGA
  
  
5443as
GGCATCCAAAGACCATCTG
380 bp
55°C
$ s = sense, as = antisense # I = inosine
Table 2
Adenoviruses, accession numbers and hosts
Adenovirus
Abbreviation
GenBank accession number
Host
Wild (Gabon)
Captive
HAdV-B of this study
     
Gorilla gorilla adenovirus B7
GgorAdV-B7
HQ292614
Western lowland gorilla
+
+
Gorilla gorilla adenovirus B8
GgorAdV-B8
HQ292615
Western lowland gorilla
+
+
Gorilla gorilla adenovirus B9
GgorAdV-B9
HQ292616
Western lowland gorilla
 
+
Gorilla gorilla adenovirus B10
GgorAdV-B10
HQ292617
Western lowland gorilla
+
 
Published HAdV-B
     
Simian adenovirus 21
SAdV-21
AC000010
Chimpanzee
 
+
Simian adenovirus 27.1
SAdV-27.1
FJ025909
Chimpanzee
 
+
Simian adenovirus 27.2
SAdV-27.2
FJ025928
Gorilla
 
+
Simian adenovirus 28.1
SAdV-28.1
FJ025914
Chimpanzee
 
+
Simian adenovirus 28.2
SAdV-28.2
FJ025915
Gorilla
 
+
Simian adenovirus 29
SAdV-29
FJ025916
Chimpanzee
 
+
Simian adenovirus 32
SAdV-32
FJ025911
Chimpanzee
 
+
Simian adenovirus 33
SAdV-33
JF025908
Chimpanzee
 
+
Simian adenovirus 35.1
SAdV-35.1
FJ025912
Chimpanzee
 
+
Simian adenovirus 35.2
SAdV-35.2
FJ025910
Bonobo
 
+
Simian adenovirus 41.1
SAdV-41.1
FJ025913
Gorilla
 
+
Simian adenovirus 41.2
SAdV-41.2
FJ025927
Gorilla
 
+
Simian adenovirus 46
SAdV-46
FJ025930
Gorilla
 
+
Simian adenovirus 47
SAdV-47
FJ025929
Gorilla
 
+
Human adenovirus B3
HAdV-B3
DQ086466
Human
  
Human adenovirus B7
HAdV-B7
AC000018
Human
  
Human adenovirus B11
HAdV-B11
AY163756
Human
  
Human adenovirus B14
HAdV-B14
AY803294
Human
  
Human adenovirus B16
HAdV-B16
AY601636
Human
  
Human adenovirus B21
HAdV-B21
AY601633
Human
  
Human adenovirus B34
HAdV-B34
AY737797
Human
  
Human adenovirus B35
HAdV-B35
AY271307
Human
  
Human adenovirus B50
HAdV-B50
AY737798
Human
  
To acquire extended sequence information of GgorAdV-B7, three additional nested PCR assays were designed (Table 1) targeting the preterminal protein (pTP) and two conserved regions at the 5'- and 3'-end of the hexon gene (Figure 1). PCRs were performed as described above, except that elongation at 72°C was for 1 min. With each primer set products of the expected size were obtained. BLAST analysis of their sequences also revealed a HAdV-B-like virus (not shown). To prove that the DPOL, pTP and hexon sequences originate from the same virus, we connected them with long-distance (LD) PCRs (Figure 1) using the TaKaRa-EX PCR system according to the instructions of the manufacturer (Takara Bio Inc., Otsu, Japan). The LD primer pairs are listed with their annealing temperatures in Table 1. Three overlapping PCR products were generated and sequenced by primer walking. A final contiguous sequence of 15637 bp was obtained spanning the genes DPOL, pTP and 52 k, the genes encoding the AdV proteins pIIIa, III (penton base), pVII, V, pX and pVI, and the hexon gene of GgorAdV-B7 (Figure 1).
Since the AdV hexon gene is an important member of the core gene set used for AdV classification [2, 3, 13, 14], we compared the available partial hexon gene of GgorAdV-B7 (2.7 kb) pair-wise with the corresponding hexon sequences of the most closely related human and chimpanzee HAdV-B viruses and all published gorilla HAdV-B viruses. The highest identity percentages were 96.6% for SAdV-35.1 (chimpanzee AdV) and 96% for HAdV-B21. The gorilla AdV SAdV-27.2, SAdV-28.2, SAdV-41.1, SAdV-41.2, SAdV-46 and SAdV-47 revealed only 87-91% identity. This closer relationship to HAdV-B21 and SAdV-35.1 was restricted to the loop-encoding regions 1 and 2 [15] as visible in an analysis with the software SIMPLOT http://​sray.​med.​som.​jhmi.​edu (Figure 2). In pair-wise comparisons of DPOL and pTP genes, GgorAdV-B7 was equally closely related to chimpanzee and gorilla HAdV-B viruses (96-99.9%). However, the penton base gene of GgorAdV-B7 showed a striking similarity (99.7%) only to that of SAdV-29 (chimpanzee AdV). Using the program mVISTA http://​genome.​lbl.​gov/​vista/​index.​shtml, the near-perfect match of the GgorAdV-B7 and SAdV-29 penton base genes over the entire gene length is clearly visible (Figure 3).
With the PhyML plug-in 2.0.1 of the Geneious Pro 5.0.4 software, phylogenetic trees were constructed on the basis of hexon, DPOL, pTP and penton base gene alignments (Figures 4). All published, completely sequenced HAdV-B viruses were included. In the hexon-based tree, GgorAdV-B7 clustered with HAdV-B21, SAdV-35.1 (chimpanzee AdV) and SAdV-35.2 (bonobo AdV) (Figure 4a). DPOL and pTP analyses placed GgorAdV-B7 into a tight cluster of gorilla and chimpanzee AdVs (Figure 4b and 4c). In the tree derived from the penton base, GgorAdV-B7 branched separately, nearly at the same position as SAdV-29 (Figure 4d). With the MrBayes 2.0.2 plug-in (Geneious Pro) or the Neighbor-Joining module of MacVector 10.6, trees with the same AdV clusters and similarly supported topology were obtained (data not shown).
The remarkably close relatedness of GgorAdV-B7 to chimpanzee AdVs (SAdV-29 and 35.1) prompted us to investigate whether gorillas naturally host GgorAdV-B7. For this purpose, we examined wild Western lowland gorillas (Gorilla. g. gorilla) from Gabon and additional captive gorillas. Fecal samples were collected from 19 individuals in a remote area with little human presence in Loango National Park, Gabon. They originated from fresh nest sites or were freshly found on gorilla paths [16]. Samples were collected using single-use gloves and preserved by drying over silica. DNA was extracted following a previously described method [17]. In addition, ten necropsy samples (spleen, liver, pancreas, lymph node, tonsil, lung, kidney, urine) and one plasma sample were collected from four deceased captive gorillas in the Zoological gardens of Berlin as well as six fecal samples from three captive gorillas in the Zoological gardens of Münster, Germany. To test for the presence of HAdV-B viruses, we set up a nested PCR (PCR Hex-loop2; Table 1) which targets flanking sequences of a hyper variable region (loop 2) in the hexon gene (Figure 1) and amplifies 380 bp. The primers were deduced from HAdV-B sequences only and not degenerated. A total of 36 gorilla samples were screened. AdV DNA was only detected in feces (5/19 wild gorilla samples and 4/6 captive gorilla samples). In total, 9/25 fecal samples were PCR-positive (36%), and the products sequenced. Most importantly, a virus apparently identical to GgorAdV-B7 was identified in a wild gorilla from Gabon. Three additional HAdV-B viruses were also detected. Two were without close similarities to any published AdV sequence. The third one was nearly identical to the gorilla AdVs SAdV-27.2 and SAdV-47, which had been originally isolated from captive individuals [12]. They were tentatively named GgorAdV-B8, -B9 and -B10.
GgorAdV-B8 was detected in two fecal samples from wild gorillas in Gabon and in one sample from Münster. Its hexon sequence revealed the highest percentage of identity (86.5%) to SAdV-21 (chimpanzee HAdV-B). GgorAdV-B9 was only amplified from captive gorillas (two fecal samples from Münster) and showed 88% identity to SAdV-29 (chimpanzee HAdV-B). The GgorAdV-B7 to -B10 Hex-loop2 sequences and closely related sequences of published gorilla, chimpanzee, bonobo and human HAdV-B viruses were subjected to phylogenetic analysis as described above. In the tree, the HAdV-B viruses segregate into several subclades with members of two, three or four host species (human, chimpanzee, bonobo and gorilla) (Figure 5). This mixed clustering was also observed upon analysis of the nearly complete hexon gene (2.7 kb; Figure 4a) with GgorAdV-B7 only. It was partially visible in the penton base tree (Figure 4d) and entirely absent from the DPOL and pTP trees (Figures 4b and 4c). In addition, the phylogenetic position of a given primate AdV frequently differed, depending on the analyzed gene (compare Figures 4a to 4d).
Taken together, these observations cannot be explained by co-speciation. Rather, they are in line with recombination and host switching. Such events have been previously discussed to be involved in the evolution of human AdV, because intra-species shuffling of penton base, fiber and hexon genes has been frequently observed [46]. In addition, a recombinant between viruses of the sub-clades HAdV-B1 and HAdV-B2 has been isolated from a captive chimpanzee [12]. Here, a close similarity of GgorAdV-B7 to SAdV-29 (complete sequence, excluding hexon loops) and to SAdV-35.1 (hexon loops) was observed (Figure 2). However, since in the loop region the nucleic acid identity between GgorAdV-B7 and SAdV-35.1 was well below 100%, it is unlikely that the existing AdVs are parent viruses in a recent recombination event giving rise to GgorAdV-B7. Rather, a more ancient one with subsequent genetic drift may have been involved or recombination with an unknown AdV, as suggested for HAdV-A18 [18].
Shuffling of genes by recombination between AdVs that naturally infect different host species (e.g., great ape and human AdVs) but under certain conditions co-infect the same host, may be an additional mechanism by which AdVs exchange genetic information. This could occur in places where contacts between humans and apes are frequent like in zoos and animal facilities. In addition, people who are involved in hunting primates and preparation of bush meat [19] are at risk to be infected. So far, infections of humans with non-human primate (NHP) AdVs have not been observed. Nevertheless, antibodies with specificity for chimpanzee HAdV-C viruses have been detected in humans from Sub-Saharan Africa and were significantly less frequent in people from the United States of America and Thailand [20]. In addition, the species HAdV-E comprises only one human serotype but more than 12 great ape serotypes. Therefore, the human HAdV-E was thought to be the result of a zoonotic transmission from chimpanzees to humans [21]. The gorilla AdV described in the present study (GgorAdV-B7) is highly similar to chimpanzee AdVs. Thus, a transmission event between chimpanzees and gorillas was possibly involved which is further indication for the potential of AdVs to jump between closely related hosts.
Very little is known about the pathogenic properties of NHP-AdV. GgorAdV-B7 was originally discovered in a group of gorillas suffering from prolonged diarrhea and self-limiting respiratory infection. Since human species B AdV have been linked to respiratory diseases [2224], an etiological association of GgorAdV-B7 with the observed respiratory symptoms is possible. However, recent studies reported the frequent shedding of AdVs in the feces of healthy captive chimpanzees and gorillas [12, 25]. Therefore, further investigations are needed.
Knowledge about the spectrum of AdV in wild great apes in general [25] is very limited. Specifically from wild gorillas, no information has been published. Although examining only a small set of samples, our findings show that AdV infecting captive gorillas can readily be found in wild animals (GgorAdV-B7; GgorAdV-B8). This is a good example of how humans may be brought into contact with new pathogens, not only locally through bushmeat hunting in regions where NHP live naturally, but also in other regions of the world where NHP are housed in zoos.
The high variety of known and novel HAdV-B viruses in great apes calls for larger studies to understand the diversity of AdVs currently circulating in African NHP as well as in local human populations. It is justified to assume that such studies will improve our insight into the zoonotic potential of adenoviruses and possibly answer the intriguing question whether AdVs of non-human primates have already contributed to the human "adeno-virosphere".

Accession numbers

The sequences reported in this study were deposited in GenBank under the accession numbers listed in Table 2.

Acknowledgements

We thank Michael Laue for electron microscopic analysis. The technical assistance of Sonja Liebmann and Cornelia Walter is kindly acknowledged. We thank the Agence Nationale des Parcs Nationaux (ANPN) and the Centre National de la Recherche Scientifique et Technique (CENAREST) of Gabon for permission to conduct research in Loango National Park. We also thank the Société pour la Conservation et le Développement (SCD) and Wildlife Conservation Society (WCS) for financial and logistical support. For sample collection from wild gorilla, we thank L. Rabanal, L. Mackaga, E. R. Guizard, N. Tagg, B. Graw, E. Fairet, M. Gregoire, L. Rankin and the other field assistants of the Loango Ape project. For DNA extraction and identification of individual samples from wild gorilla we thank M. Arandjelovic and L. Vigilant from the Max-Planck-Institute for Evolutionary Anthropology, Leipzig.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

DW and JK performed the cell culture experiments. DW, NS, JK and JH performed the molecular genetic studies. FHL, CB and MMR coordinated field work and sample collection. JH and CL sampled gorilla feces. DW, NS, JK, FHL and BE conceived of the study, and participated in its design and coordination. DW, FHL, CB, JK and BE drafted the manuscript. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Davison AJ, Benko M, Harrach B: Genetic content and evolution of adenoviruses. J Gen Virol 2003, 84: 2895-2908. 10.1099/vir.0.19497-0PubMedCrossRef Davison AJ, Benko M, Harrach B: Genetic content and evolution of adenoviruses. J Gen Virol 2003, 84: 2895-2908. 10.1099/vir.0.19497-0PubMedCrossRef
2.
Zurück zum Zitat Benkö M, Harrach B, Both G, Russel W, Adair BM, Adam E, de Jong JC, Hess M, Johnson M, Kajon A, et al.: Family Adenoviridae. In Virus Taxonomy: VIIIth Report of the International Committee on Taxonomy of Viruses. Edited by: Fauquet CM, Mayo MA, Maniloff J, Desselberger U, Ball LA. Elsevier; 2005:213-228. Benkö M, Harrach B, Both G, Russel W, Adair BM, Adam E, de Jong JC, Hess M, Johnson M, Kajon A, et al.: Family Adenoviridae. In Virus Taxonomy: VIIIth Report of the International Committee on Taxonomy of Viruses. Edited by: Fauquet CM, Mayo MA, Maniloff J, Desselberger U, Ball LA. Elsevier; 2005:213-228.
3.
Zurück zum Zitat Bailey A, Mautner V: Phylogenetic relationships among adenovirus serotypes. Virology 1994, 205: 438-452. 10.1006/viro.1994.1664PubMedCrossRef Bailey A, Mautner V: Phylogenetic relationships among adenovirus serotypes. Virology 1994, 205: 438-452. 10.1006/viro.1994.1664PubMedCrossRef
4.
Zurück zum Zitat Lukashev AN, Ivanova OE, Eremeeva TP, Iggo RD: Evidence of frequent recombination among human adenoviruses. Journal of General Virology 2008, 89: 380-388. 10.1099/vir.0.83057-0PubMedCrossRef Lukashev AN, Ivanova OE, Eremeeva TP, Iggo RD: Evidence of frequent recombination among human adenoviruses. Journal of General Virology 2008, 89: 380-388. 10.1099/vir.0.83057-0PubMedCrossRef
5.
Zurück zum Zitat Robinson CM, Rajaiya J, Walsh MP, Seto D, Dyer DW, Jones MS, Chodosh J: Computational analysis of human adenovirus type 22 provides evidence for recombination among species D human adenoviruses in the penton base gene. Journal of Virology 2009, 83: 8980-8985. 10.1128/JVI.00786-09PubMedPubMedCentralCrossRef Robinson CM, Rajaiya J, Walsh MP, Seto D, Dyer DW, Jones MS, Chodosh J: Computational analysis of human adenovirus type 22 provides evidence for recombination among species D human adenoviruses in the penton base gene. Journal of Virology 2009, 83: 8980-8985. 10.1128/JVI.00786-09PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Walsh MP, Chintakuntlawar A, Robinson CM, Madisch I, Harrach B, Hudson NR, Schnurr D, Heim A, Chodosh J, Seto D, Jones MS: Evidence of molecular evolution driven by recombination events influencing tropism in a novel human adenovirus that causes epidemic keratoconjunctivitis. PLoS One 2009., 4: 10.1371/journal.pone.0005635 Walsh MP, Chintakuntlawar A, Robinson CM, Madisch I, Harrach B, Hudson NR, Schnurr D, Heim A, Chodosh J, Seto D, Jones MS: Evidence of molecular evolution driven by recombination events influencing tropism in a novel human adenovirus that causes epidemic keratoconjunctivitis. PLoS One 2009., 4: 10.1371/journal.pone.0005635
7.
Zurück zum Zitat Kovacs G, Harrach B, Zakhartchouk AN, Davison AJ: Complete genome sequence of simian adenovirus 1: An Old World monkey adenovirus with two fiber genes. Journal of General Virology 2005, 86: 1681-1686. 10.1099/vir.0.80757-0PubMedCrossRef Kovacs G, Harrach B, Zakhartchouk AN, Davison AJ: Complete genome sequence of simian adenovirus 1: An Old World monkey adenovirus with two fiber genes. Journal of General Virology 2005, 86: 1681-1686. 10.1099/vir.0.80757-0PubMedCrossRef
8.
Zurück zum Zitat Basnight M Jr, Rogers NG, Gibbs CJ Jr, Gajdusek DC: Characterization of four new adenovirus serotypes isolated from chimpanzee tissue explants. American Journal of Epidemiology 1971, 94: 166-171.PubMed Basnight M Jr, Rogers NG, Gibbs CJ Jr, Gajdusek DC: Characterization of four new adenovirus serotypes isolated from chimpanzee tissue explants. American Journal of Epidemiology 1971, 94: 166-171.PubMed
9.
Zurück zum Zitat Kovacs GM, Davison AJ, Zakhartchouk AN, Harrach B: Analysis of the first complete genome sequence of an Old World monkey adenovirus reveals a lineage distinct from the six human adenovirus species. Journal of General Virology 2004, 85: 2799-2807. 10.1099/vir.0.80225-0PubMedCrossRef Kovacs GM, Davison AJ, Zakhartchouk AN, Harrach B: Analysis of the first complete genome sequence of an Old World monkey adenovirus reveals a lineage distinct from the six human adenovirus species. Journal of General Virology 2004, 85: 2799-2807. 10.1099/vir.0.80225-0PubMedCrossRef
10.
Zurück zum Zitat Bányai K, Esona MD, Liu A, Wang Y, Tu X, Jiang B: Molecular detection of novel adenoviruses in fecal specimens of captive monkeys with diarrhea in China. Veterinary Microbiology 2010, 142: 416-419. 10.1016/j.vetmic.2009.10.014PubMedCrossRef Bányai K, Esona MD, Liu A, Wang Y, Tu X, Jiang B: Molecular detection of novel adenoviruses in fecal specimens of captive monkeys with diarrhea in China. Veterinary Microbiology 2010, 142: 416-419. 10.1016/j.vetmic.2009.10.014PubMedCrossRef
11.
Zurück zum Zitat Kidd AH, Garwicz D, Oberg M: Human and simian adenoviruses: Phylogenetic inferences from analysis of VA RNA genes. Virology 1995, 207: 32-45. 10.1006/viro.1995.1049PubMedCrossRef Kidd AH, Garwicz D, Oberg M: Human and simian adenoviruses: Phylogenetic inferences from analysis of VA RNA genes. Virology 1995, 207: 32-45. 10.1006/viro.1995.1049PubMedCrossRef
12.
Zurück zum Zitat Roy S, Vandenberghe LH, Kryazhimskiy S, Grant R, Calcedo R, Yuan X, Keough M, Sandhu A, Wang Q, Medina-Jaszek CA, et al.: Isolation and characterization of adenoviruses persistently shed from the gastrointestinal tract of non-human primates. PLoS Pathogens 2009., 5: 10.1371/journal.ppat.1000503 Roy S, Vandenberghe LH, Kryazhimskiy S, Grant R, Calcedo R, Yuan X, Keough M, Sandhu A, Wang Q, Medina-Jaszek CA, et al.: Isolation and characterization of adenoviruses persistently shed from the gastrointestinal tract of non-human primates. PLoS Pathogens 2009., 5: 10.1371/journal.ppat.1000503
13.
Zurück zum Zitat Casas I, Avellon A, Mosquera M, Jabado O, Echevarria JE, Campos RH, Rewers M, Perez-Brena P, Lipkin WI, Palacios G: Molecular identification of adenoviruses in clinical samples by analyzing a partial hexon genomic region. J Clin Microbiol 2005, 43: 6176-6182. 10.1128/JCM.43.12.6176-6182.2005PubMedPubMedCentralCrossRef Casas I, Avellon A, Mosquera M, Jabado O, Echevarria JE, Campos RH, Rewers M, Perez-Brena P, Lipkin WI, Palacios G: Molecular identification of adenoviruses in clinical samples by analyzing a partial hexon genomic region. J Clin Microbiol 2005, 43: 6176-6182. 10.1128/JCM.43.12.6176-6182.2005PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Crawford-Miksza LK, Schnurr DP: Adenovirus serotype evolution is driven by illegitimate recombination in the hypervariable regions of the hexon protein. Virology 1996, 224: 357-367. 10.1006/viro.1996.0543PubMedCrossRef Crawford-Miksza LK, Schnurr DP: Adenovirus serotype evolution is driven by illegitimate recombination in the hypervariable regions of the hexon protein. Virology 1996, 224: 357-367. 10.1006/viro.1996.0543PubMedCrossRef
15.
Zurück zum Zitat Torres S, Chodosh J, Seto D, Jones MS: The revolution in viral genomics as exemplified by the bioinformatic analysis of human adenoviruses. Viruses 2010, 2: 1367-1381. 10.3390/v2071367PubMedPubMedCentralCrossRef Torres S, Chodosh J, Seto D, Jones MS: The revolution in viral genomics as exemplified by the bioinformatic analysis of human adenoviruses. Viruses 2010, 2: 1367-1381. 10.3390/v2071367PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Arandjelovic M, Head J, Kühl H, Boesch C, Robbins MM, Maisels F, Vigilant L: Effective non-invasive genetic monitoring of multiple wild western gorilla groups. Biological Conservation 2010, 143: 1780-1791. 10.1016/j.biocon.2010.04.030CrossRef Arandjelovic M, Head J, Kühl H, Boesch C, Robbins MM, Maisels F, Vigilant L: Effective non-invasive genetic monitoring of multiple wild western gorilla groups. Biological Conservation 2010, 143: 1780-1791. 10.1016/j.biocon.2010.04.030CrossRef
17.
Zurück zum Zitat Nsubuga AM, Robbins MM, Roeder AD, Morin PA, Boesch C, Vigilant L: Factors affecting the amount of genomic DNA extracted from ape faeces and the identification of an improved sample storage method. Mol Ecol 2004, 13: 2089-2094. 10.1111/j.1365-294X.2004.02207.xPubMedCrossRef Nsubuga AM, Robbins MM, Roeder AD, Morin PA, Boesch C, Vigilant L: Factors affecting the amount of genomic DNA extracted from ape faeces and the identification of an improved sample storage method. Mol Ecol 2004, 13: 2089-2094. 10.1111/j.1365-294X.2004.02207.xPubMedCrossRef
18.
Zurück zum Zitat Walsh MP, Seto J, Tirado D, Chodosh J, Schnurr D, Seto D, Jones MS: Computational analysis of human adenovirus serotype 18. Virology 2010, 404: 284-292. 10.1016/j.virol.2010.05.013PubMedPubMedCentralCrossRef Walsh MP, Seto J, Tirado D, Chodosh J, Schnurr D, Seto D, Jones MS: Computational analysis of human adenovirus serotype 18. Virology 2010, 404: 284-292. 10.1016/j.virol.2010.05.013PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Bennett EL, Blencowe E, Brandon K, Brown D, Burn RW, Cowlishaw G, Davies G, Dublin H, Fa JE, Milner-Gulland EJ, et al.: Hunting for consensus: Reconciling bushmeat harvest, conservation, and development policy in West and Central Africa. Conservation Biology 2007, 21: 884-887. 10.1111/j.1523-1739.2006.00595.xPubMedCrossRef Bennett EL, Blencowe E, Brandon K, Brown D, Burn RW, Cowlishaw G, Davies G, Dublin H, Fa JE, Milner-Gulland EJ, et al.: Hunting for consensus: Reconciling bushmeat harvest, conservation, and development policy in West and Central Africa. Conservation Biology 2007, 21: 884-887. 10.1111/j.1523-1739.2006.00595.xPubMedCrossRef
20.
Zurück zum Zitat Xiang Z, Li Y, Cun A, Yang W, Ellenberg S, Switzer WM, Kalish ML, Ertl HCJ: Chimpanzee adenovirus antibodies in humans, sub-Saharan Africa. Emerging Infectious Diseases 2006, 12: 1596-1599.PubMedPubMedCentralCrossRef Xiang Z, Li Y, Cun A, Yang W, Ellenberg S, Switzer WM, Kalish ML, Ertl HCJ: Chimpanzee adenovirus antibodies in humans, sub-Saharan Africa. Emerging Infectious Diseases 2006, 12: 1596-1599.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Purkayastha A, Ditty SE, Su J, McGraw J, Hadfield TL, Tibbetts C, Seto D: Genomic and bioinformatics analysis of HAdV-4, a human adenovirus causing acute respiratory disease: Implications for gene therapy and vaccine vector development. Journal of Virology 2005, 79: 2559-2572. 10.1128/JVI.79.4.2559-2572.2005PubMedPubMedCentralCrossRef Purkayastha A, Ditty SE, Su J, McGraw J, Hadfield TL, Tibbetts C, Seto D: Genomic and bioinformatics analysis of HAdV-4, a human adenovirus causing acute respiratory disease: Implications for gene therapy and vaccine vector development. Journal of Virology 2005, 79: 2559-2572. 10.1128/JVI.79.4.2559-2572.2005PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Schmitz H, Wigand R, Heinrich W: Worldwide epidemiology of human adenovirus infections. American Journal of Epidemiology 1983, 117: 455-466.PubMed Schmitz H, Wigand R, Heinrich W: Worldwide epidemiology of human adenovirus infections. American Journal of Epidemiology 1983, 117: 455-466.PubMed
23.
Zurück zum Zitat Russell WC: Adenoviruses. In Topley and Wilson's Virology 10th edition. Edited by: Mahy BWJ, Ter Meulen V, Hodder Arnold. 2005, 439-443. Russell WC: Adenoviruses. In Topley and Wilson's Virology 10th edition. Edited by: Mahy BWJ, Ter Meulen V, Hodder Arnold. 2005, 439-443.
24.
Zurück zum Zitat Gray GC, Callahan JD, Hawksworth AW, Fisher CA, Gaydos JC: Respiratory diseases among U.S. Military personnel: Countering emerging threats. Emerging Infectious Diseases 1999, 5: 379-387. 10.3201/eid0503.990308PubMedPubMedCentralCrossRef Gray GC, Callahan JD, Hawksworth AW, Fisher CA, Gaydos JC: Respiratory diseases among U.S. Military personnel: Countering emerging threats. Emerging Infectious Diseases 1999, 5: 379-387. 10.3201/eid0503.990308PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Tong S, Singh J, Ruone S, Humphrey C, Yip CCY, Lau SKP, Anderson LJ, Kaur T: Short report: Identification of adenoviruses in fecal specimens from wild chimpanzees ( Pan trogylodytes schweinfurthii ) in Western Tanzania. American Journal of Tropical Medicine and Hygiene 2010, 82: 967-970. 10.4269/ajtmh.2010.09-0668PubMedPubMedCentralCrossRef Tong S, Singh J, Ruone S, Humphrey C, Yip CCY, Lau SKP, Anderson LJ, Kaur T: Short report: Identification of adenoviruses in fecal specimens from wild chimpanzees ( Pan trogylodytes schweinfurthii ) in Western Tanzania. American Journal of Tropical Medicine and Hygiene 2010, 82: 967-970. 10.4269/ajtmh.2010.09-0668PubMedPubMedCentralCrossRef
Metadaten
Titel
A novel adenovirus of Western lowland gorillas (Gorilla gorilla gorilla)
verfasst von
Diana Wevers
Fabian H Leendertz
Nelly Scuda
Christophe Boesch
Martha M Robbins
Josephine Head
Carsten Ludwig
Joachim Kühn
Bernhard Ehlers
Publikationsdatum
01.12.2010
Verlag
BioMed Central
Erschienen in
Virology Journal / Ausgabe 1/2010
Elektronische ISSN: 1743-422X
DOI
https://doi.org/10.1186/1743-422X-7-303

Weitere Artikel der Ausgabe 1/2010

Virology Journal 1/2010 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.