key: cord-0003402-nquw7qga authors: Kuchipudi, Suresh V.; Nissly, Ruth H. title: Novel Flu Viruses in Bats and Cattle: “Pushing the Envelope” of Influenza Infection date: 2018-08-06 journal: Vet Sci DOI: 10.3390/vetsci5030071 sha: 1399e34d62b515e482ccde6c4cae500b19546909 doc_id: 3402 cord_uid: nquw7qga Influenza viruses are among the major infectious disease threats of animal and human health. This review examines the recent discovery of novel influenza viruses in bats and cattle, the evolving complexity of influenza virus host range including the ability to cross species barriers and geographic boundaries, and implications to animal and human health. Influenza viruses are known to constantly evolve and cross species barriers. The genetic diversity of influenza viruses is ever increasing with more novel influenza subtypes being discovered periodically. The purpose of this review is to provide an up-to-date overview of ecology and evolution of influenza viruses including the novel influenza viruses in bats and cattle. In addition, we discussed the growing complexity of influenza virus-host interactions and highlighted the key research questions that need to be answered for a better understanding of the emergence of pandemic influenza viruses. Influenza is among the major infectious disease problems affecting animal and human health globally. Several human influenza pandemics have been recorded since 1590 AD [1] , with the most significant of those being the "Spanish flu" of 1918, often referred to as the "mother of all pandemics" [2] . Spanish flu pandemic is believed to have affected approximately 25-30 percent of the world's population and caused more than 50-60 million human deaths globally [3] . Influenza infections in humans occur either as epidemic (seasonal or interpandemic) influenza caused by influenza A and B viruses, or as sporadic pandemic influenza caused by influenza A viruses [4] . Study of influenza pandemics has been of great interest to epidemiologists. Influenza epidemics and pandemics have been repeatedly occurring for centuries, but to date the ability to predict a pandemic has not been achieved [5] . Influenza viruses belong to the family Orthomyxoviridae (from the Greek orthos, meaning "standard, correct", and myxa, meaning "mucus"). Members of Orthomyxoviridae are characterized by virions that are either spherical or pleomorphic measuring 80-120 nm in diameter. Genomes are comprised of single-stranded negative-sense RNA that is arranged in either eight, seven or six segments depending on the genus. Classification of the historically described A, B and C influenza viruses based on antigenic differences in nucleocapsid (NP) and matrix (M1) proteins. Influenza A viruses (IAVs) are further classified into subtypes based on the antigenic properties of the external glycoproteins namely IAVs are known to infect humans and a wide variety of animals including pigs, horses, minks, seals, whales and birds [19] . Avian influenza viruses (AIVs) continue to spread, evolve and cause significant economic losses to poultry industries globally. Unlike low pathogenic avian influenza viruses (LPAIVs) that cause mild clinical signs in domestic poultry, infection of gallinaceous poultry with highly pathogenic avian influenza viruses (HPAIVs) results in severe fatal disease often causing up to 100% mortality within two-three days [20] . AIVs have also been implicated in the generation of influenza viruses that have caused all human influenza pandemics. In particular, human infection with certain contemporary Eurasian lineage HPAI H5N1 viruses frequently caused severe disease with a case fatality rate of up to 50% [21] . Ability to infect a wide range hosts is a key contributing factor to the complex and seemingly expanding genetic diversity of IAVs. It is now well established that IAVs infect domestic pets such as dogs and cats, adding to the list of host species that could potentially expose humans to influenza viruses. An H3N8 equine influenza virus in 1999 and an avian virus-like H3N2 strain around 2005 or 2006 were transmitted to dogs, and these canine influenza viruses have been circulating in the U.S. dog population ever since [22] . After the first report of pandemic H1N1 influenza A virus (pH1N1) infection of cats in Italy in 2011 [23] , more than 500 cats became infected with influenza subtype H7N2 in animal shelters in New York, NY, USA during 2016-2017 [24] . While transmission of these animal viruses to humans has not yet been documented, the close contact of many humans with pet dogs and cats presents an increased risk of opportunity for such a host jump. Transmission of IAVs between animals and humans is bidirectional such that humans are infected by animal influenza viruses and human influenza viruses have been shown to transmit to animals. In particular, studies established that domestic pets are susceptible to infection by human influenza viruses. For example, dogs experimentally inoculated with a human seasonal H3N2 or pandemic (pdm) H1N1 (2009) showed nasal shedding and seroconversion. However, dogs inoculated with influenza B virus did not exhibit virus shedding or seroconversion highlighting that interspecies transmissibility is a key feature of IAVs and not IBVs [25] . Aquatic birds such as ducks, gulls, and shorebirds have historically been identified as the primary reservoir of all the known IAV subtypes [26] . Recently, Northwest Atlantic gray seals were suggested as an endemically infected wild reservoir population for diverse influenza viruses [27] . Arguably, influenza virus host range could be much broader than currently known, with additional reservoirs that are yet to be revealed. Prior to 2011, 16 antigenically different HA (H1-H16) and nine different NA (N1-N9) types had been described, all of which were found in the aquatic bird reservoir. However, the number of HA and NA types have now expanded after the identification of two novel influenza-like viruses in fruit bats. The first was in a frugivorous yellow-shouldered bat (Sturnira lilium) in Guatemala [28] . Soon after, a second virus was found in the flat-faced fruit bat (Artibeus planirostris) in Peru [29] . The newly identified bat flu viruses are genetically distinct from all previously known IAVs and hence are designated as novel subtypes namely H17N10 and H18N11 [29] . Serological surveys showed wide spread prevalence of H17/H18-specific antibodies among bat populations in Central and South America [29] . Finding novel bat-influenza viruses is not surprising as bats represent 24% of all known mammalian species [30] and have been known to be natural reservoirs of many deadly zoonotic RNA viruses including Ebola virus [31] [32] [33] . It is believed that bats have the capacity to harbor more influenza virus genetic diversity than all the other mammalian and avian species combined [29] . Notably, little yellow-shouldered bats in Central America have been proposed as a potential sylvatic mammalian reservoir of influenza [28] . Discovery of novel bat IAVs raises numerous questions including the host range evolution of IAVs and the role of bats in evolution of IAVs. The potential for the newly discovered bat IAVs to reassort with previously known IAV types is being explored and providing novel information about the IAV life cycle. Bat flu viruses have been reconstructed by reverse genetics using synthetic DNA which has allowed for more functional understanding of these viruses [34] . Multiple experimental studies have shown that reassortment of bat IAVs and conventional IAVs does not occur under experimental conditions [35] [36] [37] . This is partly explained by functional differences between bat IAVs and conventional IAVs. IAV nonstructural protein NS1 is a multifunctional protein that plays a crucial role in evading host immune responses and serves as a key virulence factor [38, 39] . Unlike the NS1 of conventional IAVs, bat NS1 fails to bind to the host p85β, a regulatory subunit of the cellular metabolism-regulating enzyme phosphoinositide 3-kinase (PI3K) [40] . In addition, the surface glycoprotein HA and NA genes of bat IAVs only show low nucleotide sequence identity with those of the conventional IAVs [28, 29] . Notably, it has been found that the bat IAV hemagglutinin (HA) and neuraminidase (NA) proteins lack the receptor binding activity that are characteristic of the conventional IAV [28, 41] . HA of bat IAVs does not bind to the classical avian (SAα2,3-Gal) or human (SAα2,6-Gal) IAV receptors highlighting the possibility of unique entry mechanisms that are yet to be identified [42] . Consequently, it was found that bat IAVs initiate infection at the basolateral membrane of cells unlike conventional IAVs which preferential initiate infection on the apical surface of cells [34] . These findings highlight that there are major evolutionary constraints to bat IAVs to effectively reassert with conventional IAVs and/or infect other species including humans. While the zoonotic potential of bat IAVs is not yet fully established and continues to be a focus of ongoing research, bats have been found to be susceptible to infection by conventional IAVs. A recent study found serological evidence of IAV H9 subtype infection in 30% of frugivorous bats tested in Africa [43] . A key source of IAV genetic diversity could come from the replication of IAVs in a non-native host species that initiate evolution of new virus variants [44] . The receptors for previously known IAVs are sialic acids (SA) on host cells [45] . Consequently, the expression of these receptors on host cells is a key determinant of the ability of IAVs to infect a host [46] . AIVs preferentially bind to SA receptors that are linked to galactose by an α 2,3 linkage (SAα2,3-Gal), while human and classical swine influenza viruses show preference to α2,6 linked SAs (SAα2,6-Gal). It is widely believed that hosts that co-express both SAα2,3-Gal and SAα2,6-Gal receptors could support reassortment of IAVs and hence play a major role in the evolution of IAVs [45, 47] . We have demonstrated abundant co-expression of both SAα2,3-Gal and SAα2,6-Gal receptors that are compatible with avian and human IAV binding in respiratory and digestive tracts of little brown bats, the most widespread bat species in North America [48] . The potential for bats to support infection by multiple conventional IAV types and thus serve as a source of novel genetic variants remains to be fully explored. Although influenza viruses infect humans and a wide range of animals and birds, cattle were never considered to be susceptible to influenza virus infection. However, a novel influenza virus has recently been identified in several animals including swine, cattle, sheep, and goats. The virus was first isolated as an influenza C-like virus from pigs with respiratory illness in Oklahoma, USA, in 2011 [49, 50] . The virus was subsequently classified as influenza D virus (IDV), and the virus has now been reported from many countries including United States, France [51] , Italy [52] , China [53] , Japan [54] Ireland [55] and countries in North and West Africa [56] . The International Committee on Taxonomy of Viruses (ICTV), which is responsible for developing, refining, and maintaining a universal virus taxonomy, has recently released revised classification of Orthomyxoviridae [57] . In earlier classification, Orthomyxoviruses belonged to any one of the five genera: Influenzavirus A, Influenzavirus B, Influenzavirus C, Thogotovirus and Isavirus [58] . However, the most recent classification of Orthomyxoviridae includes eight genera and nine species (Table 1) . Genus Alphainfluenzavirus comprises the species influenza A virus, Betainfluenzavirus comprises the species influenza B virus, Gammainfluenzavirus comprises the species influenza C virus, and Delatinfluenzavirus comprises the species IDV [58] . Like other influenza viruses, IDVs possess a negative-sense single-stranded RNA genome. However unlike IAVs that contain 8 segments, IDVs comprise seven genomic segments that are predicted to encode nine proteins, including glycoprotein hemagglutinin-esterase fusion (HE), polymerases PB2, PB1, and P3, nucleoprotein, matrix protein (M1 and CM2), and nonstructural proteins (NS1 and NEP) [59] . IDVs use 9-O-acetylated sialic acid as their cellular receptor on host cells such as ICVs [60] . It is well established based on several epidemiological studies that cattle are the primary reservoirs of IDVs [50, 59] . In addition, IDVs have also been isolated from a range of animals including pigs, sheep, goat, horses and camelids. While the precise role of IDVs in clinical disease in animals is not yet fully investigated, their role in causing respiratory infections in cattle has been implied. Two recent studies carried out metagenomic characterizations of the virome associated with bovine respiratory disease in feedlot cattle and found correlation of IDV presence with Bovine respiratory disease (BRD) clinical signs, raising exciting new prospects for understanding and combatting this complicated disease [61, 62] . BRD complex is one of the major diseases affecting the cattle industry in the USA and around the world. Productivity losses due to BRD are estimated to be $23.60 per calf with an annual economic impact of more than one billion dollars to the U.S. cattle industry [61] . BRD is associated by multiple pathogens and accounts for approximately 70-80% of the morbidity in the USA [63] and 84.5-99.9% of the morbidity in Mexican feedlot cattle. BRD results in the use of widespread therapeutics and antibiotics in feedlots, which increasingly raises public health concerns of promoting antibiotic resistance [64, 65] . Pathophysiology of BRD involves complex interactions between host, pathogen, environment and management factors. In feedlot cattle, BRD is initiated by viral infection followed by stress due to travel which is typically followed by a secondary infection by resident bacteria [66] . Viral infection can cause increased susceptibility to secondary bacterial infections by either immunosuppression or by damaging the epithelium of upper airways and injuring lung parenchyma which facilitates the migration of bacterial pathogens and colonization of the lower respiratory tract. Depending on several factors, the clinical outcome of BRD can be variable; however, higher morbidity and mortality are observed in the event of mixed viral and bacterial infections [67] . Many viral pathogens have been implicated in BRD, which include bovine viral diarrhea virus, bovine herpesvirus 1, bovine respiratory syncytial virus and bovine parainfluenza 3. Experimental studies in calves with IDV showed damage that results in the induction of inflammation in the trachea. IDV could be a significant player in BRD and could facilitate coinfections with other bovine pathogens [59] . In addition to the animal health implications of IDVs, a recent study found IDV antibodies in 34 out of 35 persons that had contact with cattle and only 2 out of 11 that did not have any exposure to cattle [68] . The results of this study raised the possibility that IDV could be relevant from a public health stand point and that it could pose a zoonotic risk to cattle-exposed workers. With much still to be characterized about the new IDV species, exploring its impact on human an animal health through epidemiological studies will be vital to understanding spread of this virus. Emerging and novel zoonotic infections often result from pathogens jumping from their original host into novel host species [69] . The host range evolution of mammalian viruses typically involves more closely related hosts [70, 71] . In particular, RNA viruses with broad host range are more likely to jump between distantly related species [70, 72, 73] . A key determinant in the host range evolution of a virus is the mechanism of viral entry used by the viruses. Analysis of 64 human viruses revealed that the viruses that use receptors that are highly conserved in their amino acid sequence across species have the broadest host range [74] . The ability of viruses to bind to an alternative receptor is sometimes key in species jumping. AIVs need to change to preferentially bind to SAα-2,6-Gal receptor to efficiently transmit between humans. In experimental studies, it was shown that the shift from SAα-2,3-Gal to SAα-2,6-Gal binding requires four mutations for a HA of HPAI H5N1 viruses [75] . However, several newly emerged H5N1 AIVs in Egypt have been found to have acquired the human receptor SAα-2,6-Gal binding ability during their emergence in birds [76] . Influenza A, B, C and D viruses have varying susceptible host range ( Figure 1) . Notably, the newly discovered IDVs have the widest host range after IAVs. Further, humans and pigs are susceptible to infection by all four types of influenza viruses. With the extensive host range that continues to grow and the zoonotic potential, influenza viruses remain a major challenge to epidemiologists. Owing to their tremendous potential to affect animal and human health, there is a need to carry out in-depth and comprehensive studies to unravel the ecological complexity of influenza virus host range evolution. In particular, we feel that researchers should focus on answering the key questions, "what is the role of bats in the ecology and evolution of IAVs?", "are IDVs involved in the epidemic influenza infections in people?", and "are birds susceptible to infection by IDVs?" Vet. Sci. 2018, 5, x FOR PEER REVIEW 6 of 10 need to carry out in-depth and comprehensive studies to unravel the ecological complexity of influenza virus host range evolution. In particular, we feel that researchers should focus on answering the key questions, "what is the role of bats in the ecology and evolution of IAVs?", "are IDVs involved in the epidemic influenza infections in people?", and "are birds susceptible to infection by IDVs?" A history of influenza Influenza: The mother of all pandemics The origin and virulence of the 1918 "Spanish" influenza virus Reviewing the History of Pandemic Influenza: Understanding Patterns of Emergence and Transmission A history of influenza Influenza: The mother of all pandemics The origin and virulence of the 1918 "Spanish" influenza virus Reviewing the History of Pandemic Influenza: Understanding Patterns of Emergence and Transmission Non coding extremities of the seven influenza virus type C vRNA segments: Effect on transcription and replication by the type C and type A polymerase complexes Domestic pigs are susceptible to infection with influenza B viruses Epidemiology and pathogenesis of influenza Selection and analysis of antigenic variants of the neuraminidase of N2 influenza viruses with monoclonal antibodies Antigenic and genetic characteristics of swine-origin 2009 A(H1N1) influenza viruses circulating in humans Avian-to-human transmission of the PB1 gene of influenza A viruses in the 1957 and 1968 pandemics Compatibility among polymerase subunit proteins is a restricting factor in reassortment between equine H7N7 and human H3N2 influenza viruses Selective packaging of the influenza A genome and consequences for genetic reassortment The genome-packaging signal of the influenza A virus genome comprises a genome incorporation signal and a genome-bundling signal Interaction network linking the human H3N2 influenza A virus genomic RNA segments A supramolecular assembly formed by influenza A virus genomic RNA segments An in vitro network of intermolecular interactions between viral RNA segments of an avian H5N2 influenza A virus: Comparison with a human H3N2 virus A functional sequence-specific interaction between influenza A virus genomic RNA segments Viral determinants of influenza A virus host range Experimental infection of chickens, ducks and quails with the highly pathogenic H5N1 avian influenza virus Mammalian innate resistance to highly pathogenic avian influenza H5N1 virus infection is mediated through reduced proinflammation and infectious virus release Influenza virus reservoirs and intermediate hosts: Dogs, horses, and new possibilities for influenza virus exposure of humans Influenza A pandemic (H1N1) 2009 virus outbreak in a cat colony in Italy Characterization of a Feline Influenza A(H7N2) Virus Canine susceptibility to human influenza viruses (A/pdm 09H1N1, A/H3N2 and B) Highly pathogenic avian influenza virus infection in chickens but not ducks is associated with elevated host immune and pro-inflammatory responses Prevalence of influenza A virus in live-captured North Atlantic gray seals: A possible wild reservoir A distinct lineage of influenza A virus from bats New world bats harbor diverse influenza A viruses Bats and their virome: An important source of emerging viruses capable of infecting humans Middle East respiratory syndrome coronavirus in bats, Saudi Arabia Bats as "special" reservoirs for emerging zoonotic pathogens Bats are a major natural reservoir for hepaciviruses and pegiviruses Synthetically derived bat influenza A-like viruses reveal a cell type-but not species-specific tropism An infectious bat-derived chimeric influenza virus harbouring the entry machinery of an influenza A virus Characterization of uncultivable bat influenza virus using a replicative synthetic virus Pathogenicity of modified bat influenza virus with different M genes and its reassortment potential with swine influenza A virus Influenza virus sequence feature variant type analysis: Evidence of a role for NS1 in influenza virus host range restriction Conformational plasticity of the influenza A virus NS1 protein Unexpected Functional Divergence of Bat Influenza Virus NS1 Proteins Structural and functional characterization of neuraminidase-like molecule N10 derived from bat influenza A virus Bat-derived influenza hemagglutinin H17 does not bind canonical avian or human receptors and most likely uses a unique entry mechanism Serological evidence of influenza A viruses in frugivorous bats from Africa Influenza A virus polymerase is a site for adaptive changes during experimental evolution in bat cells Differences in influenza virus receptors in chickens and ducks: Implications for interspecies transmission Endocytosis of influenza viruses. Microbes Infect Comparative distribution of human and avian type sialic acid influenza receptors in the pig Avian and human influenza virus compatible sialic acid receptors in little brown bats Isolation of a novel swine influenza virus from Oklahoma in 2011 which is distantly related to human influenza C viruses Characterization of a novel influenza virus in cattle and Swine: Proposal for a new genus in the Orthomyxoviridae family Influenza D virus in cattle Detection of Influenza D Virus among Swine and Cattle Influenza D Virus Infection in Herd of Cattle Influenza D Virus in Cattle Serologic Evidence for Influenza C and D Virus among Ruminants and Camelids Pathogenesis of Influenza D Virus in Cattle An Open Receptor-Binding Cavity of Hemagglutinin-Esterase-Fusion Glycoprotein from Newly-Identified Influenza D Virus: Basis for Its Broad Cell Tropism Metagenomic characterization of the virome associated with bovine respiratory disease in feedlot cattle identified novel viruses and suggests an etiologic role for influenza D virus A metagenomics and case-control study to identify viruses associated with bovine respiratory disease Where have we been, where are we now, and where do we want to go? Anim A ten-year (2000-2009) study of antimicrobial susceptibility of bacteria that cause bovine respiratory disease complex-Mannheimia haemolytica, Pasteurella multocida, and Histophilus somni-In the United States and Canada Metaphylactic antimicrobial therapy for bovine respiratory disease in stocker and feedlot cattle Review of BRD pathogenesis: The old and the new Board-invited review: Recent advances in management of highly stressed, newly received feedlot cattle Serologic evidence of exposure to influenza D virus among persons with occupational contact with cattle The evolution and genetics of virus host shifts Phylogeny and geography predict pathogen community similarity in wild primates and humans A tale of two phylogenies: Comparative analyses of ecological interactions Emerging pathogens: The epidemiology and evolution of species jumps Host range and emerging and reemerging pathogens Human viruses: Discovery and emergence Experimental adaptation of an influenza H5 HA confers respiratory droplet transmission to a reassortant H5 HA/H1N1 virus in ferrets Acquisition of human-type receptor binding specificity by new H5N1 influenza virus sublineages during their emergence in birds in Egypt Acknowledgments: Authors acknowledge the support of Penn State Animal Diagnostic laboratory for providing resources and access needed for the preparation of this review. The authors declare no conflict of interest.Acknowledgments: Authors acknowledge the support of Penn State Animal Diagnostic laboratory for providing resources and access needed for the preparation of this review. The authors declare no conflict of interest.