Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Nov 1.
Published in final edited form as: Trends Neurosci. 2023 Sep 6;46(11):953–970. doi: 10.1016/j.tins.2023.08.002

The neuropathogenesis of highly pathogenic avian influenza H5Nx viruses in mammalian species including humans

Lisa Bauer 1,*, Feline F W Benavides 1,*, Edwin J B Veldhuis Kroeze 1, Emmie de Wit 2, Debby van Riel 1,#
PMCID: PMC10591965  NIHMSID: NIHMS1929831  PMID: 37684136

Abstract

Circulation of highly pathogenic avian influenza (HPAI) H5Nx viruses of the A/Goose/Guangdong/1/96 lineage in birds regularly causes infections of mammals, including humans. In many mammalian species, infections are associated with severe neurological disease, a unique feature of HPAI H5Nx viruses compared to other influenza A viruses. Here, we provide an overview of the neuropathogenesis of HPAI H5Nx virus infections in mammals, centered on three aspects: neuroinvasion, neurotropism, and neurovirulence. We focus on in vitro studies, as well as studies on naturally or experimentally infected mammals. Additionally, we discuss the contribution of viral factors to the neuropathogenesis of HPAI H5Nx virus infections, and the efficacy of intervention strategies to prevent neuroinvasion or the development of neurological disease.

Keywords: central nervous system, neuroinflammation, microbiology, viral encephalitis, emerging viruses, zoonoses

Emergence and circulation of H5Nx viruses

Highly pathogenic avian influenza (HPAI) H5Nx viruses of the A/Goose/Guangdong/1/96 (Gs/Gd) lineage emerged more than 25 years ago and were first isolated from domestic geese in 1996 [1]. Shortly after, the first human infection was documented [2] and since then a total of 868 human cases have been detected of which 457 were fatali. The phylogenetic tree of the Gs/Gd lineage is based on the sequence of the trimeric surface protein hemagglutinin (HA)ii [3]. The extensive circulation and continuous evolution of H5N1 viruses led to a diversification of the HA protein, resulting in multiple clades and subclades, of which subclade 4.3.3.2b is currently spreading worldwide via wild birds [4], [5]. Additionally, reassortment (see Glossary) events have occurred which led to the exchange of non-HA gene segments between viruses. This resulted in local circulation of for example H5N6 or H5N8 viruses, which are all descendants of the ancestral Gs/Gd isolate that emerged in 1996 [6]–[9]. In this review, we refer to the Gs/Gd lineage viruses as HPAI H5Nx viruses.

Prior to 2005, HPAI H5Nx viruses were predominately circulating in poultry species in Asia, with incidental spillovers to migratory birds resulting in local outbreaks without sustained transmission within wild birds year-round [9]. Since 2021, HPAI H5Nx viruses, specifically from clade 2.3.4.4b, are circulating continuously in wild birds with intercontinental detections in Asia, Africa, Europe, North America and South America, resulting in mass mortalities in different wild bird species [10], [11]. In addition, HPAI H5Nx viruses are frequently detected in mammalian species that feed on sick or dead infected birds. So far, mammal-to-mammal transmission seems rare, but has been suggested in 2003 among tigers [12], [13] and recently in minks [14] and sea lions [15].

Neuropathogenesis of H5Nx viruses

A unique feature of HPAI H5Nx viruses is their ability to cause severe neurological disease in birds and mammals, a feature rarely observed for other influenza A viruses. Neurological complications are the most common clinical manifestations in naturally infected mammals, although one could argue that this may be due to the ease of observation of neurological versus respiratory symptoms. However, studies from experimentally infected mammals show that neurological complications occur more frequently after HPAI H5Nx virus infection compared to infection with low pathogenic avian influenza (LPAI), seasonal or pandemic influenza A viruses [16], [17]. To date, neurological disease has been reported in many mammalian species including humans, foxes, cats, tigers, stone martens, harbor porpoises, common seals, gray seals, ferrets, mice, pikas, and minks (Table 1) [11]. In this review, we discuss current knowledge on the neuropathogenesis of HPAI H5Nx virus infections in mammals, focusing on three aspects: neuroinvasion, neurotropism and neurovirulence. Although we focus on mammals, we summarize the neuropathogenic potential of these HPAI H5Nx viruses in birds in Box 1. Lastly, we outline the role of viral factors and how these contribute to neuropathogenicity of HPAI H5Nx viruses in mammals, and possible intervention strategies and treatment options in humans.

Table 1.

Distribution of sialic acids in the CNS of various mammals.

Species α(2,3) sialic acids (MAA) α(2,6) sialic acids (SNA) References
Areas Cell type Areas Cell type
Mouse Olfactory bulb: Yes
Hippocampus: Yes
Thalamus: Yes
Cerebellum: Yes
Pons: No
Medulla oblongata: No
N, VE, E Olfactory bulb: Yes
Hippocampus: Yes
Thalamus: Yes
Cerebellum: Yes
Pons: Yes
Medulla oblongata: Yes
N, G, VE, E [131]
Cerebrum: No
Cerebellum: Yes
- Cerebrum: No
Cerebellum: Yes
- [132]
Substantia nigra: Yes
Hippocampus: Yes
Cerebellum: Yes
- Substantia nigra: No
Hippocampus: No
Cerebellum: No
- [133]
Human Cerebral cortex: Yes
Hippocampus: Yes
Brain stem: Yes
Cerebellum: Yes
N, G, VE, E Cerebral cortex: Yes
Hippocampus: Yes
Brain stem: Yes
Cerebellum: Yes
N, G, VE, E [131]
- N, VE - VE [134]
Pig - N - VE [135]
Cat Cerebellum: Yes Cerebellum: Yes [136]
Dog - VE - VE [137]

It should be noted that experimental variation between different lectin suppliers is observed and needs critical evaluation [138]. Abbreviations: N = neurons, G = glial cells, VE = vascular endothelial cells, E = ependymal cells, - = data not available.

Box 1: The neuropathogenic potential of HPAI H5 viruses in birds.

Neurological complications are regularly observed in birds infected with HPAI H5Nx viruses. Many species develop neurological signs, including chickens and turkeys (Galliformes), ducks and geese (Anseriformes) and raptors (Falconiformes) [170]–[177]. The neuropathogenicity of HPAI H5Nx viruses differs among avian species. Infection starts in the respiratory tract and/or gastrointestinal tract, after which the virus can spread to other tissues including the CNS [177]. The route of neuroinvasion is not well studied in birds. In gallinaceous poultry HPAI H5Nx viruses have a strong endotheliotropism, which is not observed in wild birds like mallard ducks [177]. Infected damaged endothelial cells may lead to hemorrhages, edema, congestion, and thrombosis [177], and form a likely route of hematogenous virus spread to the CNS in gallinaceous poultry species. The route of neuroinvasion in bird species without evident endotheliotropism like ducks remains to be unraveled. Inside the CNS, various cells of the CNS including ependymal cells, glial cells and neurons may become infected, demonstrating the virus’ neurotropism. Infected cells produce progeny virus and become necrotic, which triggers infiltration of inflammatory cells, gliosis, hemorrhages and edema [177], [178], consistent with an encephalitis. Systemic spread and multiorgan virus infection usually induces similar acute necrohemorrhagic lesions, most notably within lungs, heart, pancreas, and liver. If evident, macroscopic CNS lesions may consist of malacia, hemorrhages and edema [177], [178]. Neurological signs commonly reported include, head twitching, ataxia, tremors, torticollis, and opisthotonus [177], [178]. The degree of neurovirulence of HPAI H5Nx viruses depends on the virus and the infected bird species. For example, natural infection in farmed domestic ducks was shown to result in more severe neurological disease compared to wild mallard ducks [178]. Furthermore, HPAI H5Nx infection in raptors like buzzards, falcons, and sea eagles is associated with severe, often fatal, neurological disease [172], [173], [175], [176]. Since the global spread of HPAI H5Nx viruses, a wider range bird species has been infected, which resulted in mass mortality in sensitive bird species [179]. Further, the carcasses of infected birds form a source of infection for opportunistic scavenging terrestrial and aquatic carnivores.

Neuroinvasion

Neuroinvasion refers to the ability of a virus to enter the peripheral or central nervous system (CNS) [18]. Influenza A viruses initially infect cells within the respiratory tract, and from there, can spread to the CNS. Possible pathways include virus transport within or along cranial nerves (Figure 1A & B) or hematogenous spread after which it may cross the blood-brain-barrier (BBB) or blood-cerebral spinal fluid-barrier (BCSF). Cranial nerves that innervate the mammalian respiratory tract include the olfactory nerve (CN I; sensory fibers) and trigeminal nerve (CN V; sensory and motor fibers) in the nasal cavity, the facial nerve (CN VII; sensory, parasympathetic and motor fibers) and glossopharyngeal nerve (CN IX; sensory and motor fibers) in the upper respiratory tract, and the vagus nerve (CN X; sensory, parasympathetic and motor fibers) in the lower respiratory tract. Evidence suggests that HPAI H5Nx viruses can use the olfactory [16], [19]–[30], trigeminal [22]–[24], [27], [30], [31], facial [28], [31], vestibulocochlear [19], [22], [27] the vagus [22]–[24] and upper thoracic sympathetic nerves [23] to enter the CNS in mammals, as virus antigen is detected in the soma and/or axons of sensory neurons of these nerves (Figure 1C).

Figure 1. Routes of neuroinvasion for HPAI H5Nx viruses.

Figure 1.

(A) The olfactory pathway includes the olfactory mucosa which contains olfactory sensory neurons. The ciliated neurons extend into the nasal cavity and their axons pass through the cribriform plate and terminate in the olfactory bulb in the brain. (B) The respiratory pathway includes the respiratory mucosa. Nerve endings of sensory neurons reside behind the tight-junctions of epithelial cells and are not in direct contact with the environment. Their cell bodies are found in ganglia. (C) Presence of H5N1 virus nucleoprotein antigen is shown in the olfactory pathway and the respiratory pathway in experimentally infected ferrets. Images adjusted from [30]. Figure created with Biorender.

The nasal cavity is lined by respiratory and olfactory mucosa. The surface ratio of respiratory and olfactory mucosa differs between mammalian species. In humans, it is estimated that around 1.25% of the nasal mucosa consists of olfactory mucosa (2–10 cm2) [32], [33], while in mice this is around 47% [34]. The olfactory mucosa is responsible for odor recognition and comprises olfactory sensory neurons (OSN), sustentacular cells, microvillar cells, basal cells, olfactory ensheathing cells, and cells of the Bowman’s glands [32]. The dendrites of the bipolar OSN have cilia that are directly exposed to the environment (Figure 1A) [35], [36]. Neuroinvasion along the olfactory nerve starts by virus infection of cells within the olfactory mucosa. Several studies in mice and ferrets have shown that HPAI H5Nx viruses replicate efficiently in the olfactory mucosa with virus antigen in all cell types including OSN [20], [29], [30]. Infection of OSN can result in anterograde transport of virus through the cribriform plate of the ethmoid bone into the olfactory bulb, the brain structure involved in olfaction. In the olfactory bulb, axons of the OSN synapse in glomeruli with mitral cells and periglomerular cells in the glomerular layer, which have been found infected in experimentally inoculated ferrets [20], [29]. Alternatively, viruses could diffuse through continuous, fluid-filled channels created by olfactory ensheathing cells along the olfactory nerve that are filled with CSF and end up in the meninges [25]. Efficient replication of HPAI H5Nx viruses in the olfactory mucosa likely contributes to this propensity for neuroinvasion, as seasonal and pandemic influenza viruses replicate less efficiently in the olfactory mucosa and are less frequently associated with neuroinvasion along the olfactory nerve [16], [37]. Transmission of HPAI H5N1 virus along the olfactory nerve does not result in a genetic bottleneck [30].

Within the respiratory mucosa, nerve endings of the sensory neurons are located directly below the tight junctions of the respiratory epithelial cells, and unlike OSN endings, are not in direct contact with the environment (Figure 1B). The nerve fibers of the trigeminal nerve innervate amongst others the respiratory mucosa of the nasal cavity, the nasopharynx, the sinuses, and the palate. CNS invasion along the trigeminal nerve has been observed in experimentally inoculated mice and ferrets, where virus antigen was detected in the trigeminal ganglion [20], [23], [24], [29], [38]. Evidence for neuroinvasion of HPAI H5Nx viruses along the facial nerve is based on the detection of virus antigen in the facial nucleus and solitary nucleus in experimentally inoculated mice and ferrets [22], [28], [39]. The vestibulocochlear pathway has been implicated in CNS invasion in ferrets and mice. In ferrets, HPAI H5Nx virus antigen was detected in epithelial cells of the Eustachian tube [27], and in the cochlea and vestibulocochlear nerve [22]. In intranasally inoculated mice, virus antigen was detected in vestibulocochlear nuclei [22]. Neuroinvasion along the vagus nerve, of which sensory nerve terminals are widely distributed throughout the lower respiratory tract, has been shown in mice [28], [38], [40].

Hematogenous spread of HPAI H5Nx viruses could result in subsequent CNS entry through the BBB or BCSFB, but there is little evidence that HPAI H5Nx viruses invade the CNS via this route. HPAI H5Nx viruses can spread to the circulation (viremia) in both humans and experimentally inoculated animals [41]–[44]. In contrast, viremia is not commonly observed during seasonal influenza virus infections in mammals [43], [45], [46]. To our knowledge, there is currently no evidence for trans- or paracellular transport of cell free or cell-associated HPAI H5Nx virus over the BBB or BCSFB. However, in intragastrically inoculated cats and naturally infected foxes, HPAI H5Nx viruses have been shown to infect few endothelial cells in the CNS, from where viruses could spread across the BBB [47], [48]. Of note, although in poultry and black swans endothelial cell infection with HPAI H5Nx viruses is a common observation, endothelial infection in mammals appears to be rare.

Neurotropism

Neurotropism is the ability of a virus to infect cells of the CNS and replicate in them [18]. Once the virus invades the CNS, a variety of cell types in the brain can become infected, such as neurons, glial cells (astrocytes, microglia, oligodendrocytes, and ependymal cells), choroid plexus cells, neural endothelial cells, and pericytes. The infection efficiency of HPAI H5Nx viruses varies among the different CNS cell types.

The surface protein HA facilitates attachment to and entry into susceptible cells. The receptor binding properties of HA determine in part the cell tropism. Avian viruses, for example, preferentially bind to α(2,3) sialic acids (SIAs), whereas human influenza viruses recognize α(2,6) SIAs (reviewed in [49]). While the presence of SIAs in the respiratory tract is well studied [50]–[52], their presence in different anatomical locations of the mammalian CNS is largely uncharacterized. The CNS has a high SIA content, and several studies suggest the presence of both α(2,3)- and α(2,6)-linked SIAs in mammalian species such as mice, pigs, cats, dogs and humans (Table 1). SIA distribution in the CNS of ferrets, one of the most widely used animal models for influenza research, has not been investigated, to our knowledge.

In vitro and in vivo, HPAI H5Nx viruses can infect many different CNS cell types. In vitro studies showed that HPAI H5Nx viruses infect and replicate in neuron- and astrocyte-like cells [53], [54], in primary mouse astrocytes and microglia [55], [56], and in different human induced pluripotent stem cell (hiPSC)-derived cell types such as neural progenitor cells, neurons and astrocytes [57]. These in vitro studies show efficient infection of CNS cells, based on increasing viral titers in the supernatant over time. In experimentally inoculated ferrets, mice and pikas, and naturally infected red foxes, cats, tigers, stone martens, harbor porpoises, common seals and gray seals, both neurons and glial cells were infected with HPAI H5Nx virus, based on the detection of virus antigen (Table 2 & 3) [13], [20], [23]–[28], [58]–[73]. Occasionally, infected ependymal, choroid plexus and meningeal cells were observed [20], [27], [62], [63], [66], [72], [74], [75]. The detection of virus antigen in different anatomical locations, and the isolation of infectious virus from them (Figure 2) indicate that HPAI H5Nx viruses replicate efficiently within the CNS of mammals [16], [76]. However, the replication efficiency and distribution within the CNS seems to be strain dependent. For instance, while A/HK/483/97 is known to disseminate throughout the CNS, A/HK/486/97 does not exhibit the same level of spread, despite both strains being able to invade the CNS [77]. The neurotropism in humans is poorly studied as post-mortem CNS samples are often not collected for various reasons. However, in the few cases described, influenza virus antigen was detected in neurons and glial cells in various areas of the brain such as the cerebral cortex, hippocampus, midbrain and cerebellum (Table 4) [78]–[80].

Table 2.

Experimentally HPAI H5Nx virus-inoculated mammals in which neuroinvasion and/or neurovirulence has been observed.

Strain Species Neuroinvasion Neurotropism Neurovirulence References
Inflammatory CNS lesions Neurological
A/Hong Kong/156/97 Macaques Yes* No No No No [139]
Mice Yes* N, G - Yes - [24], [60]
A/Hong Kong/483/97 Mice Yes, ON, TN, VN N, G, S, E Yes Yes Yes [23], [24], [60], [69], [77], [121], [140]
Ferret Yes, ON No - Yes Yes [31], [69], [140], [141]
A/Hong Kong/486/1997 Mice Yes* - - - No [69], [140]
Ferret Yes* - - Yes Yes [69], [140], [141]
A/chicken/Hong Kong/YU822.2/2001 Mice Yes* - - Yes Yes [142]
A/chicken/Hong Kong/YU562/2001 Mice No - - - - [142]
A/pheasant/Hong Kong/FY155/2001 Mice Yes* - - - Yes [142]
A/chicken/Hong Kong/FY150/2001 Mice Yes* - - - Yes [142]
A/chicken/Hong Kong/NT873.3/2001 Mice Yes* - - - Yes [142]
A/Hong Kong/213/03 Ferret Yes* - - - - [70]
A/Chicken/Indonesia/7/03 Mice No - - - No [26], [69]
Ferret No - - - -
A/Chicken/Vietnam/NCVD/8/2003 Mice No - - - - [69]
A/Chicken/Korea/ES/2003 Mice No - - - No [69]
Ferret Yes* - - - No
A/Vietnam/1194/2004 Cats Yes* N, G, E - Yes No [62]
A/Vietnam/1203/04 Mice Yes, ON, VN, FN N, G, E Yes Yes Yes [26], [28], [66], [69], [90]
Ferrets Yes, ON, VON, H N, G, E Yes Yes [19], [26], [27], [31], [69], [70], [76]
Hamsters Yes* - - - - [76]
A/Vietnam/1204/04 Mice Yes* - - - No [69]
Ferrets Yes* N - Yes Yes
A/Vietnam/UT3062/04 Ferret Yes* - - - - [76]
Hamsters Yes* - - - -
A/Thailand/16/2004 Mice Yes* - - - No [69]
Ferrets Yes* - - Yes No
A/Thailand/SP/83/2004 Mice No - - - No [69]
Ferrets Yes* - - No No
A/Thailand/Kan/353/2004 Ferret Yes* - - Yes Yes [69]
A/Chicken/Vietnam/NCVD/31/2004 Mice No - - - No [69]
Ferrets Yes* - - No No
A/birds/Qinghai/07/04 Mice Yes* - - Yes No [143]
A/Whooper swan/Mongolia/244/05 Mice Yes* N.S. - Yes - [26]
Ferrets Yes* - - - No
A/Muscovy duck/Vietnam/ 209/05 Mice Yes* N.S. - Yes Yes [26]
Ferret - - - - No
A/Vietnam/JP36–2/05 Ferret Yes* N - Yes Yes [70]
A/Indonesia/5/2005 Ferrets Yes, ON N, G, E - Yes Yes [20], [25]
A/goose/Krasnoozerskoye/627/05 Mice Yes* - Yes - - [144]
A/turkey/Suzdalka/12/05 Mice Yes* - - - - [144]
A/Turkey/65–596/06 Ferret Yes* - - No No [70]
A/duck/Tuva/01/06 Mice Yes* - - - - [144]
A/chicken/Krasnodar/123/06 Mice Yes* - - - - [144]
A/chicken/Reshoty/02/06 Mice Yes* - - - - [144]
A/whooper swan/Germany/R65-1/2006 Red fox Yes* N, G - Yes No [67]
A/ great black-headed gull/Qinghai/1/2009 Pika Yes* N - Yes - [58]
A/Vietnam/HN36285/2010 Cats Yes* N.S. - Yes No [145]
Dogs No - - No No
A/crow/India/02CA01/2012 Mice Yes* - - Yes - [64]
A/black-headed gull/Netherlands/29/2017 (H5N6) Ferret Yes, ON, TN N, G, E - Yes No [146]

All virus strains are H5N1 viruses unless otherwise indicated. Abbreviations: * = route of neuroinvasion is not specified, ON = olfactory nerve, TN = trigeminal nerve, VN = vagus nerve, VON = vestibulocochlear nerve, FN = Facial nerve, H = hematogenous. Neurotropism is defined as positive signal IHC/ISH in defined cell type. N.S = cell type not specified, N = neuron, G = glial cells, S = Schwann cells, E = ependymal cells. - = data not available.

Table 3.

Naturally HPAI H5Nx virus-infected mammals in which neuroinvasion and/or neurovirulence has been observed.

Year(s) and location of infection Species Neuroinvasion Neurotropism Neurovirulence References
CNS lesions Neurological signs
2003 - Thailand Tiger, leopard - - Yes - [12]
2004 - Thailand Cat Yes* N Yes Yes [68]
2004 - Thailand Dog No - No - [147]
2004 - Thailand Tiger, leopard Yes* N Yes Yes [13]
2005 - Vietnam Civet Yes* N Yes Yes [148]
2006 - Germany Cat Yes* - - - [149]
2006 - Germany Stone marten Yes* N, G Yes Yes [71]
2013 - China Tiger Yes* - N.A. Yes [150]
2014/2015 - China Tiger - - Yes Yes [151]
2016/2017 - South Korea (H5N6) Cat Yes* N, G, E Yes Yes [75]
2020 - United Kingdom (H5N8) Seals Yes* N Yes Yes [72]
Red fox N, E No
2021 - The Netherlands Red fox Yes* N.A. - Yes [81]
2021 - Germany (H5N8) Harbor seal Yes* N, G Yes - [152]
2021/2022 - Finland Otter Yes* Yes Yes [153]
Red fox No -
Lynx N.A.
2021/2022 - The Netherlands Red fox Yes* N, G Yes Yes [61]
2021/2022 - The Netherlands Red fox, polecat, badger, otter Yes* N Yes Yes [82]
2022 - Sweden Porpoise Yes* N, G, E Yes Yes [73]
2022 - Canada Red fox Yes* N.A. Yes Yes [48]
Mink, skunk - -
2022 - USA Seals - - - Yes [154]
2022 - Spain Mink - - - Yes [14]
2023 - Peru Sea lion - - - Yes [155]
2023 - Chile Sea lion - - Yes Yes [15]
2023 - USA Dolphin Yes* N Yes - [156]

All isolated viruses are H5N1 viruses unless otherwise indicated.

* =

route of neuroinvasion is not specified. Neurotropism is defined as positive signal IHC/ISH in defined cell type.

Abbreviations: N.S = cell type not specified, N = neuron, G = glial cells, S = Schwann cells, E = ependymal cells. - = data not available.

Figure 2. Anatomical locations with confirmed HPAI H5Nx virus detection in the mammalian brain.

Figure 2.

HPAI H5Nx viruses are detected in different anatomical locations within the brain. Here, all locations in which virus antigen has been detected in experimentally or naturally infected mammals are projected on a schematic overview of the human brain. Figure created with Biorender.

Table 4.

Human cases of HPAI H5Nx virus infection in which neuroinvasion and/or neurovirulence has been observed.

Year(s) and location of infection Age, sex Neuroinvasion Neurotropism Neurovirulence Outcome References
CNS lesions Neurological symptoms
1997 - Hong Kong 13F No - Yes Yes Fa [157], [158]
25F - Fa
24F - R
2003/2004 - Thailand 6M Yes - Yes# No Fa [159], [160]
2004 - Vietnam 4M Yes - - Yes Fa [161]
2004/2005 - Vietnam, Thailand, Cambodia 5 patients* - - - Yes - [162]
2005 - Indonesia 8F - - - Yes Fa [163]
21M R
2005 - Cambodia 4 patients* - - - Yes - [164]
2005 - Ho Chi Minh City 1 patient* - - - Yes - [162]
2005 - China 24F - - - Yes Fa [79]
35M Yes N No
2005/2008 - China 4 patients* - - - Yes - [165]
2005/2006 - Indonesia 7 patients* - - - Yes - [162]
2005/2006 - Turkey 1 patient* - - - Yes - [166]
2006 - Azerbaijan, Turkey 7 patients* - - - Yes - [162]
2006/2007 - Egypt 19 patients* - - - Yes - [162]
2008 - China 42M Yes N, G Yes No Fa [78]
2012 - China 2M Yes - Yes Yes R [167]
2013 - Canada 28F Yes - Yes Yes Fa [168]
2022 - China (H5N6) 6F Yes - Yes Yes R [169]

All isolated viruses are H5N1 viruses unless otherwise indicated. Neurotropism is defined as positive signal IHC/ISH in defined cell type. Abbreviations: F = female, M = male, * = age and sex unknown. N.S = cell type not specified, N = neuron, G = glial cells, S = Schwann cells, E = ependymal cells. - = data not available, # = also induction of proinflammatory cytokines, Fa = fatal, R = recovered.

Taken together, after neuroinvasion, HPAI H5Nx viruses can infect and replicate efficiently in various CNS cell types in different mammalian species. Depending on the route of neuroinvasion, various cells become exposed after which the virus possibly disseminates to other regions of the CNS. The mechanism of HPAI H5Nx virus transmission between cells of the CNS is not well studied, but it has been shown that transport of HPAI H5Nx virus can occur through transaxonal transport within axons of primary dorsal root ganglia neurons, yet transsynaptic transmission of viruses was not shown [28].

Neurovirulence

Neurovirulence refers to the ability of the virus infection to cause damage in the CNS that contributes to the development of clinical disease, independent of the neuroinvasive and neurotropic potential of the virus [18]. The neurovirulence of HPAI H5Nx virus infection is predominantly observed and studied in cases where virus entered and spread through the CNS. However, HPAI H5Nx virus infections might also trigger neurological complications in the absence of neuroinvasion. Neurological signs in HPAI H5Nx virus infected mammals include ataxia, tremors, convulsions, paralysis and seizures, and are observed in many mammalian species including cats, tigers, and foxes [13], [48], [61], [68], [75], [81], [82]. Whether there are differences in the (neuro)pathogenesis among infected mammals is currently unknown (Table 2 & 3).

In naturally infected mammals, neurological signs following HPAI H5Nx virus infection are often observed, which might relate to the attention these animals attract due to atypical behaviors like ataxia, heightened aggression, or lack of fleeing behavior (Table 3). In humans, neurological symptoms vary considerably, ranging from mild headache to severe symptoms like seizures and convulsions (Table 4). In vivo studies show that HPAI H5Nx neurovirulence is determined in part by the route and dose of inoculation, as well as virus strain and infected animal species (Table 2). For example, inoculation with A/Muscovy-duck/Vietnam/209/05 resulted in neurological signs in mice, but not in ferrets [26]. However, what proportion of infections results in severe neurological signs, and how this specifically differs among mammalian species and virus isolates is currently unknown (Figure 3).

Figure. 3. Disease pyramid of the neurological signs associated with HPAI H5Nx virus infections in mammals.

Figure. 3.

(A) Factors that influence neuroinvasion and neurotropism of HPAI H5Nx viruses. (B) The distribution of neurological disease and symptom severity following HPAI H5Nx virus infection is largely unknown. The neurovirulence of the virus can be broadly divided into four categories, ranging from no symptoms to severe symptoms. Figure created with Biorender.

The neurovirulence of HPAI H5Nx virus infection is associated with virus entry into and spread throughout the CNS, as well as the resulting damage and inflammation. Infection of CNS cells can result in cell death (via necrosis and/or apoptosis) and incite an inflammatory response which can lead to a dysregulated neuronal homeostasis. Inflammation is typically characterized by induction of pro-inflammatory cytokines and chemokines, infiltration of inflammatory cells, activation of glial cells (gliosis), and edema. Malacia and hemorrhage may be present in severe inflammatory lesions. Such acute lesions in HPAI H5Nx virus-infected mammals typically co-localize with the presence of virus antigen and are consistent with a diagnosis of viral encephalitis or meningo-encephalitis [25], [30].

Cell death of HPAI H5Nx virus-infected CNS cells has been observed in vitro and in vivo. In vitro, apoptosis was induced by HPAI H5Nx viruses in human astrocyte-like cells, and primary mouse microglia and astrocytes [54], [56]. Apoptosis might be triggered by upregulation of genes involved in interferon-α/β (IFN) signaling, Toll-like/RIG-I-like receptor signaling pathways, or elevation of internal Ca2+ in human astrocyte-like cells [54]. In vivo, HPAI H5Nx virus infection triggers neuronal degradation and cell death [20], [29].

Infection of various brain cells with HPAI H5Nx virus elicits an immune response that can cause dysregulation of the brain homeostasis. These immune-based mechanisms can trigger influx of immune cells to the site of inflammation or dysregulated neuronal plasticity by changing synapse structure and function [83], [84]. In vitro HPAI H5N1 infection of neuron-like cell lines, hiPSC-derived neural models or primary CNS cells results in the induction of proinflammatory cytokines such as type-I IFN, type-III IFN, IFN γ-induced protein 10 (IP-10), interleukin-1β (IL-1β), IL-6, IL-8 and tumor necrosis factor-α (TNF-α) [54], [56], [57], [85], [86]. In vivo, experimental infection of ferrets with influenza A viruses induced TNF-α, IL-6 and IL-8 in endothelial cells, neurons and glial cells in the CNS [87]–[89]. Cell and tissue damage co-localizes with an influx of inflammatory cells, which may include monocytes, macrophages, lymphocytes, plasma cells, and neutrophils [20], [24], [29]. Furthermore, HPAI H5Nx virus infection elicits a prominent IP-10 response in the CNS [28], [90], which is a chemoattractant for immune cells such as monocytes, macrophages and dendritic cells and weakens the integrity of the BBB [91]. However, how each of these chemokines and cytokines contribute to the development of neuroinflammation or clinical disease is not fully understood, even though it is known that for example type I IFN can cause headaches [92].

Additionally, influenza A virus infections can disturb the brain homeostasis through alterations in hippocampal neuron morphology and neuronal connectivity [93]. This can be attributed to either virus replication within CNS cells but also to local immune responses. Cytokines such as IL-1β, IL-6 and TNF-α emerge as important cytokines that disrupt the baseline physiology of synapses [84]. Unlike IL-6, which dampens neural activity in the CA1 region of the hippocampus [94], IL-1β and TNF-α can enhance excitatory as well as inhibitory neurotransmission through decreasing or enhancing the flow of Na+, K+ or Ca2+ ions [95]–[100]. All three cytokines have been linked to seizures, and memory and learning deficits in various models of epilepsy, multiple sclerosis and neurodegeneration [101]–[104]. HPAI H5Nx virus infection in an astrocyte-like cell line changed expression profiles of genes associated with neuroactive ligand-receptor interaction and internal Ca2+, important for neuron-to-neuron signaling transduction and synaptic plasticity (see Glossary) [54]. Non-structural protein 1 (NS1) of HPAI H5Nx virus interacts with postsynaptic density protein 95 (PSD95), which reduces nitric oxide (NO) levels [105]. In vivo studies in mice described accumulation of α-synuclein in the olfactory bulb, hippocampus, locus coeruleus, and solitary nucleus up to 90 days post infection [106]. These data together suggest that HPAI H5Nx viruses disrupt neural homeostasis which might contribute to the development of neurodegenerative diseases.

To summarize, HPAI H5Nx viruses, unlike seasonal or pandemic influenza A viruses, are highly neurovirulent in many mammalian species. The neurovirulence caused by the high neuroinvasive and neurotropic potential of HPAI H5Nx viruses, together with the induction of systemic cytokines can contribute to the development of severe neurological signs, including behavioral changes such as lack of fleeing behavior and aggression. Several studies suggest that HPAI H5Nx virus infection can result in milder CNS complications when the virus does not enter the CNS or when virus replication within the CNS is quickly controlled, but how this modulates behavioral outcome or neurological symptoms is not well understood. The exact types and frequencies of neurological complications associated with HPAI H5Nx infection as well as the temporal and spatial kinetics of the neurovirulence of HPAI H5Nx viruses are not fully understood (see Figure 3 and Outstanding questions).

Outstanding questions.

  • Many mammalian species infected with HPAI H5Nx viruses develop neurological disease, but it remains unknown how often infection results in neuroinvasion and neurological disease, and whether this differs among mammalian species. What is the frequency of virus invasion into the CNS in different mammalian species, including humans?

  • Neurological disease observed in HPAI H5Nx virus-infected mammals is often severe or even fatal. This might only reflect ‘the tip of the iceberg’, and it is likely that less severe neurological disease occurs unnoticed in at least some mammalian species. What is the spectrum of neurological disease in different mammalian species?

  • Differences in neuropathogenesis are observed among different isolates of the HPAI H5Nx Gs/Gd lineage viruses. Under similar experimental conditions, some viruses cause neurological disease in mammals, whereas others do not. Which viral factors and phenotypic characteristics are necessary, or essential, for the neuroinvasive, neurotropic and neurovirulent potential of HPAI H5Nx viruses?

  • HPAI H5Nx viruses show a broad neurotropism infecting different CNS cells, resulting in a pro-inflammatory response in the CNS. What cell types and which cell-intrinsic and cell-autonomous mechanisms contribute to CNS disease?

  • Individual studies have revealed that vaccination or antiviral therapies can potentially interfere with the neuroinvasion into, and spread of HPAI H5Nx viruses through the CNS, but this has not been studied comprehensively. Can intervention strategies be used to prevent or treat neurological disease caused by HPAI H5Nx viruses?

Viral factors that contribute to neuropathogenesis

There is currently limited understanding of viral factors that contribute to the neuropathogenesis of HPAI H5Nx viruses. Even though several studies identified viral factors that contribute to the replication efficiency in mammals, or transmission among them, these rarely investigate the contribution of these factors to neuropathogenesis (see Outstanding questions). However, it is known that the neuropathogenesis of HPAI H5Nx viruses cannot be attributed to one single viral protein.

Sialic Acid Binding Preference

HPAI H5Nx viruses bind preferentially to α(2,3) SIA, but whether the recognition of specific SIA contributes to neuroinvasiveness and neurotropism is currently unknown. Viruses that recognize either α(2,3) or α(2,6) linked SIA attach to the olfactory mucosa of humans and ferrets. Furthermore, ferret studies have shown that both viruses that recognize α(2,3)- or α(2,6)-linked SIA are able to replicate within the olfactory mucosa, suggesting that SIA preference is not a major determinant in the ability to infect and replicate in cells of the olfactory mucosa [20], [107]. Within the olfactory mucosa, highly poly-sialylated OSNs protrude through the submucosa into the environment where influenza A virus particles can directly attach and bind to (Figure 1). However, which SIA are specifically expressed on cilia of OSN is unknown [108]. The nerve endings of other cranial nerves are mainly in contact with respiratory epithelial cells that express α(2,6) SIA, and are therefore predominantly target cells for seasonal and pandemic influenza viruses [62], [109]. With regard to the neurotropism, HPAI H5N1 virus (which binds α(2,3) SIA), attached and replicated more efficiently in neuron and astrocyte-like cell lines compared to viruses that recognized α(2,6)-linked SIA (pH1N1 and H3N2) [53]. These data suggest that the recognition of α(2,3)-linked SA might not play an important role in the neuroinvasive potential of HPAI H5Nx viruses, but that it might contribute to neurotropism.

Multibasic Cleavage site

The HA proteins of HPAI viruses, including HPAI H5Nx viruses, contain a multibasic cleavage site (MBCS). The MBCS allows HA cleavage and activation by ubiquitously expressed proteases. This is in contrast to the monobasic cleavage site of LPAI, seasonal and pandemic influenza viruses that are cleaved by specific trypsin-like serine proteases such as human airway trypsin-like protease (HAT), transmembrane serine protease 2 (TMPRSS2), TMPRSS4 or matriptases, which are not ubiquitously expressed [110]–[112]. The presence of the MBCS coincides with the systemic spread of HPAI viruses in birds, especially poultry species, and likely also in mammals [113]. In ferrets, the MBCS is critical for the neuroinvasion of HPAI H5N1 virus along the olfactory nerve. Deletion of the MBCS from H5N1/Indonesia/2005 virus, prevented virus invasion into the CNS in ferrets, likely because replication within the olfactory mucosa was less efficient compared to H5N1/Indonesia/2005 with a MBCS [20]. However, the MBCS alone is not sufficient for neuroinvasion, as the insertion of a MBCS in a seasonal H3N2 virus did not increase the neuroinvasive potential in ferrets [114]. In mice and ferrets, virus spread to the CNS was not consistently observed after inoculation with different HPAI H7 viruses [115].

The presence of a MBCS in HPAI H5Nx viruses also contributes to the neurotropism. In vitro studies showed that both, H5N1/Indonesia/2005 with and without the MBCS, replicate in neuron-like cell lines and primary mouse cortex cells, although H5N1/Indonesia/2005 without the MBCS replicated less efficiently, despite replicating equally efficiently in MDCK cells [53]. Together, these studies indicate that the presence of MBCS contributes to the neuroinvasive and neurotropic potential of HPAI viruses, but the presence of a MBCS alone is not sufficient for neuroinvasion nor neurotropism.

Polymerase Genes

The replication efficiency of influenza A viruses in mammalian cells is largely dependent on the viral polymerase genes PB1, PB2 and PA. A genetic screen has shown that mutations that increase polymerase activity in HPAI H5Nx viruses, like a single amino acid substitution in PB1 (N105S), or PB2 (E192K, Q591K, E627K, D701N or D701V) increase replication in the nasal turbinates which correlated with higher virus titers in the brains of inoculated mice [116]. How these amino acid substitutions directly relate to the neuropathogenesis of HPAI H5Nx virus is not known. The E627K amino acid substitution in PB2 is one of the best studied in the light of mammalian adaptation, but in mice and ferrets the E627K substitution also increases the neuroinvasive potential [55], [69], [117]. Furthermore, the acquisition of 627K, or other amino acid substitutions that increase polymerase activity (PB1–117G and 635T), within the CNS has been observed in naturally infected foxes and experimentally inoculated ferrets [30], [61], [82]. Together, these studies suggest that an increased polymerase activity might influence the neuroinvasive and neurotropic potential, possibly through more efficient replication.

Other factors contributing to neuropathogenesis

PB1-F2.

Within the PB1 gene segment, an alternative start codon gives rise to the PB1-F2 protein. Amino acid position 66 in PB1-F2 was shown to be associated with increased neuroinvasion and neurotropism. In experimentally inoculated mice and ferrets amino acid substitution N66S in PB1 increases the neuropathogenicity of HPAI H5N1 virus [118], most likely through suppression of the type-I IFN response [119].

NS1.

The non-structural protein NS1 antagonizes the host innate immune responses through interacting with multiple host proteins [120]. One of these host proteins is cleavage and polyadenylation specificity factor 30 (CPSF30), a cellular factor required for processing of cellular pre-mRNA [121]. NS1 amino acid residues at position 103 and 106 are critical for a stronger interaction with CPSF30 which was associated with a faster spread of HPAI H5N1 virus especially in CNS cells [122]. In vitro evidence suggests that overexpression of NS1 derived from HPAI H5N1 but not pH1N1 virus, affects synaptic plasticity in rat neurons [123] most likely by interacting with PSD-95, a scaffold protein localized at the postsynaptic density [105].

Intervention strategies and treatment options

In view of the sensitivity of H5Nx viruses to neuraminidase inhibitors, and the lack of approved vaccines, the CDCiii and the WHOiv recommend usage of neuraminidase inhibitors. However, the efficacy of this approach in humans to prevent or treat neurological complications is poorly characterized. Hints on the efficacy of potential therapeutic intervention strategies, such as vaccination or antivirals, to prevent neurological disease can be found in preclinical studies. Mouse and ferret studies, for instance, have shown that vaccination with a homologous HPAI H5 vaccine prevented or reduced HPAI H5Nx virus neuroinvasion via the olfactory nerve [29]. Virus replication in the olfactory mucosa was less abundant or even absent in vaccinated ferrets, suggesting that vaccination limits virus spread to the CNS along the olfactory nerve [29]. Whether protective responses induced by the vaccine are effective, and how broad they are, is not well known and needs further investigation. Of note, a heterologous H3N2 vaccination did not prevent HPAI H5N1 virus neuroinvasion into the CNS in ferrets [25]. A comprehensive comparison of different vaccination strategies should reveal the optimal strategy to prevent CNS invasion by different HPAI H5Nx virus isolates. Combining virological, immunological and pathological analyses in these studies could provide important insights into the correlates of protection.

Antiviral treatment studies revealed differences in efficacy among studies, dose regimens and virus isolates. In one ferret study, prophylactic oseltamivir did not prevent HPAI H5N1 virus replication in the olfactory mucosa, neuroinvasion or virus spread throughout the CNS [29]. However, other studies revealed a reduction in virus titers or spread in the brain and neurological signs [124], [125], and in mice, baloxavir marboxil did reduce virus titers in the CNS [126]. Future studies should reveal the potential of antivirals to prevent CNS invasion or reduce virus replication within the CNS, as well as efficient dosing regimens. These studies should assess the bioavailability of the different antivirals in different anatomical locations in the CNS, as for example oseltamivir, one of the most frequently used antivirals against influenza, has low bioavailability in the CNS [127], [128].

Altogether, comprehensive studies should reveal the efficacy of vaccines and antivirals to prevent invasion and virus replication in the CNS (see Outstanding questions). As the majority of studies so far focus on the reduction of virus replication in the lungs, future studies should include analyses of the olfactory mucosa and different anatomical parts of the CNS. These studies have a broad impact in the context of pandemic preparedness and should complement the focus on treatment of HPAI H5Nx virus infections, as neurological disease has been associated with past influenza A virus pandemics.

Concluding remarks

The global spread of HPAI H5Nx viruses of the Gs/Gd lineage in wild birds is a unique event that not only affects birds but also mammals. Since 2021, sustained circulation of HPAI H5Nx viruses in wild bird populations has resulted in a tremendous increase of infected bird species as well as transmission events to mammalian species. Transmission to mammals, presumably after feeding on sick or dead birds, and the high frequency of neurological diseases in these mammals raises concerns. In humans, HPAI H5Nx viruses have also been associated with neurological disease, but the exact risk for neurological disease after infection with currently circulating HPAI H5Nx viruses, and whether this is lower compared to other mammals, is not known (see Outstanding questions).

In mammals, the development of HPAI H5Nx virus-associated neurological disease can occur without any evidence of respiratory disease. For example, virus replication within the olfactory mucosa can result in neuroinvasion and spread throughout the CNS, without virus replication in other parts of the upper or lower respiratory tract. This comes with diagnostic challenges, as respiratory samples can test negative, despite efficient virus replication in the CNS. Therefore, it is important to increase awareness among veterinarians, health care workers, and neurologists to be vigilant about HPAI H5Nx viruses causing neurological disease, without the presence of overt respiratory disease [43].

The spectrum of CNS disease associated with HPAI H5Nx infection in different mammalian species remains unclear (Figure 3). It is likely that only those cases that develop severe disease from a HPAI H5Nx virus infection are documented with the current surveillance systems, and that HPAI H5Nx virus infections that result in less severe disease are missed. A recent study in the Netherlands revealed that HPAI H5N1 virus infections were detected in carnivores without obvious neurological signs or morbidity, with serological evidence for infection in about 20% of the study population [129]. Similarly, limited human data suggest that only few cases of severe encephalitis occur, but milder neurological complications are frequently detected (Table 4). Although the disease pyramid for neurological disease is influenced by virus strain, virus dose, exposure route and the species infected, it is important to acquire more insight into the diversity of CNS complications, as well as the frequency of these complications (Figure 3). Furthermore, it is currently unclear which host factors influence the neuropathogenesis, and thus contribute to differences among species. Finally, long-term effects of HPAI H5Nx virus infection need to be studied. It is likely that encephalitis will result in long-lasting neurological deficits, like those observed after West Nile and SARS-CoV-2 virus infections [18], [130].

A comprehensive understanding of the phenotypic and genotypic characteristics of HPAI H5Nx viruses that contribute to the development of neurological disease is urgently needed. Several viral-intrinsic features have been associated with the neuropathogenesis of influenza A viruses, such as the recognition of α(2,3)-linked SIAs, the presence of a MBCS and an increased polymerase activity. However, none of these are solely responsible for the neuroinvasive, neurotropic and neurovirulent potential of HPAI H5Nx viruses. In vivo and in vitro models are indispensable to identify viral factors that contribute to, or are essential for the neuroinvasion, neurotropism and neurovirulence. Scalable in vitro hiPSC-derived neural models are particularly useful tools to investigate virus-neural cell interactions in detail, as well as to characterize the neurotropic and neurovirulent potential of newly emerging H5Nx and other viruses in a human-related model system.

To conclude, HPAI H5Nx viruses are unique among influenza A viruses in their neuroinvasive potential, their efficient replication within the CNS, and their potential to cause severe CNS disease in mammals. With the continuous circulation of HPAI H5Nx viruses worldwide, mammals, including humans, are at risk of being infected. Although sustained transmission among mammals infected with HPAI H5Nx viruses is rare, it is critical to monitor the spread of this virus. A collaborative One Health approach is required to gain insights into the frequency and the full spectrum of neurological disease in humans and animals, and identify viral factors that contribute to the neuropathogenicity of HPAI H5Nx viruses. Finally, awareness should be raised among animal and human health care workers regarding neurological complications associated with HPAI H5Nx viruses, and the potential of these neurological manifestations to occur in the absence of respiratory disease.

Highlights.

  • HPAI H5Nx viruses can cause neurological complications in many mammalian species, including humans.

  • Neurological disease induced by HPAI H5Nx viruses in mammals can manifest without clinical respiratory disease.

  • HPAI H5Nx viruses are more neuropathogenic in mammals compared to other influenza A viruses.

  • Severe neurological disease in mammals is related to the neuroinvasive and neurotropic potential of HPAI H5Nx viruses.

  • Cranial nerves, especially the olfactory nerve, are important routes of neuroinvasion for HPAI H5Nx viruses.

  • HPAI H5Nx viruses have a broad neurotropic potential, and can efficiently infect and replicate in various CNS cell types.

  • Vaccination and/or antiviral therapy might in part prevent neuroinvasion and neurological disease following HPAI H5Nx virus infection, although comprehensive studies in this area are lacking.

Acknowledgments

L.B. is supported by The Netherlands Organization for Scientific Research (XS contract number OCENW.XS22.2.045) and by a grant 2023 from the European Society of Clinical Microbiology and Infectious Diseases (Europäische Gesellschaft für klinische Mikrobiologie und Infektionskrankheiten) (ESCMID). E.D.W. is supported by the Intramural Research Program of NIAID, NIH. D.V.R. is supported by fellowships from The Netherlands Organization for Scientific Research (VIDI contract 91718308) the European Union’s Horizon Europe FARM2FORK research and innovation programme Kappa Flu, grant number 101084171.

Glossary

Neuroinvasiveness

The ability of a virus to enter either the peripheral or central nervous system

Neurotropism

The ability of a virus to infect and replicate in cells of the nervous system. Cells of the nervous system include neurons, glial cells (e.g. astrocytes, oligodendrocytes, oligodendrocyte precursor cells, microglia), meningeal cells, choroid plexus cells, and cells of the neurovascular system (such as endothelial cells and pericytes)

Neurovirulence

The ability of a virus infection to cause damage in the CNS that contributes to the development of clinical disease of the nervous system independently of the ability of the virus to invade the CNS and infect cells of the CNS

Reassortment

Reassortment is a characteristic of multi-segmented RNA viruses and refers to the exchange of viral gene segments in cells co-infected with at least two different viruses to produce virus progeny with a novel genome combination

Synaptic plasticity

Synaptic plasticity refers to the ability of synaptic connections to strengthen or weaken over time, which is an important neurophysiological process supporting learning, memory, and other cognitive functions

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Declaration on interest

The authors declare no competing interests.

References

  • [1].Xu X, Subbarao null, Cox NJ, en Guo Y, ‘Genetic characterization of the pathogenic influenza A/Goose/Guangdong/1/96 (H5N1) virus: similarity of its hemagglutinin gene to those of H5N1 viruses from the 1997 outbreaks in Hong Kong’, Virology, vol. 261, nr. 1, pp. 15–19, aug. 1999, doi: 10.1006/viro.1999.9820. [DOI] [PubMed] [Google Scholar]
  • [2].Subbarao K e.a., ‘Characterization of an avian influenza A (H5N1) virus isolated from a child with a fatal respiratory illness’, Science, vol. 279, nr. 5349, pp. 393–396, jan. 1998, doi: 10.1126/science.279.5349.393. [DOI] [PubMed] [Google Scholar]
  • [3].WHO/OIE/FAO H5N1 Evolution Working Group, ‘Toward a unified nomenclature system for highly pathogenic avian influenza virus (H5N1)’, Emerg Infect Dis, vol. 14, nr. 7, p. e1, jul. 2008, doi: 10.3201/eid1407.071681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Lee D-H, Bertran K, Kwon J-H, en Swayne DE, ‘Evolution, global spread, and pathogenicity of highly pathogenic avian influenza H5Nx clade 2.3.4.4’, J Vet Sci, vol. 18, nr. S1, pp. 269–280, aug. 2017, doi: 10.4142/jvs.2017.18.S1.269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Cui P e.a., ‘Global dissemination of H5N1 influenza viruses bearing the clade 2.3.4.4b HA gene and biologic analysis of the ones detected in China’, Emerg Microbes Infect, vol. 11, nr. 1, pp. 1693–1704, dec. 2022, doi: 10.1080/22221751.2022.2088407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Lewis NS e.a., ‘Emergence and spread of novel H5N8, H5N5 and H5N1 clade 2.3.4.4 highly pathogenic avian influenza in 2020’, Emerg Microbes Infect, vol. 10, nr. 1, pp. 148–151, dec. 2021, doi: 10.1080/22221751.2021.1872355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [7].Gu W e.a., ‘Novel H5N6 reassortants bearing the clade 2.3.4.4b HA gene of H5N8 virus have been detected in poultry and caused multiple human infections in China’, Emerg Microbes Infect, vol. 11, nr. 1, pp. 1174–1185, dec. 2022, doi: 10.1080/22221751.2022.2063076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].King J e.a., ‘Highly pathogenic avian influenza virus incursions of subtype H5N8, H5N5, H5N1, H5N4, and H5N3 in Germany during 2020–21’, Virus Evol, vol. 8, nr. 1, p. veac035, 2022, doi: 10.1093/ve/veac035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].King J, Harder T, Conraths FJ, Beer M, en Pohlmann A, ‘The genetics of highly pathogenic avian influenza viruses of subtype H5 in Germany, 2006–2020’, Transbound Emerg Dis, vol. 68, nr. 3, pp. 1136–1150, mei 2021, doi: 10.1111/tbed.13843. [DOI] [PubMed] [Google Scholar]
  • [10].Kuiken T, Fouchier RAM, en Koopmans MPG, ‘Being ready for the next influenza pandemic?’, Lancet Infect Dis, vol. 23, nr. 4, pp. 398–399, apr. 2023, doi: 10.1016/S1473-3099(23)00117-2. [DOI] [PubMed] [Google Scholar]
  • [11].European Food Safety Authority e.a., ‘Avian influenza overview December 2022 - March 2023’, EFSA J, vol. 21, nr. 3, p. e07917, mrt. 2023, doi: 10.2903/j.efsa.2023.7917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Keawcharoen J e.a., ‘Avian influenza H5N1 in tigers and leopards’, Emerg Infect Dis, vol. 10, nr. 12, pp. 2189–2191, dec. 2004, doi: 10.3201/eid1012.040759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Thanawongnuwech R e.a., ‘Probable tiger-to-tiger transmission of avian influenza H5N1’, Emerg Infect Dis, vol. 11, nr. 5, pp. 699–701, mei 2005, doi: 10.3201/eid1105.050007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Agüero M e.a., ‘Highly pathogenic avian influenza A(H5N1) virus infection in farmed minks, Spain, October 2022’, Euro Surveill, vol. 28, nr. 3, p. 2300001, jan. 2023, doi: 10.2807/1560-7917.ES.2023.28.3.2300001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Gamarra-Toledo V e.a., ‘Mass Mortality of Marine Mammals Associated to Highly Pathogenic Influenza Virus (H5N1) in South America’. bioRxiv, p. 2023.02.08.527769, 3 maart 2023. doi: 10.1101/2023.02.08.527769. [DOI] [Google Scholar]
  • [16].van den van den Brand JMA e.a., ‘Comparison of Temporal and Spatial Dynamics of Seasonal H3N2, Pandemic H1N1 and Highly Pathogenic Avian Influenza H5N1 Virus Infections in Ferrets’, PLOS ONE, vol. 7, nr. 8, p. e42343, aug. 2012, doi: 10.1371/journal.pone.0042343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Kuiken T en Taubenberger JK, ‘Pathology of human influenza revisited’, Vaccine, vol. 26 Suppl 4, pp. D59–66, sep. 2008, doi: 10.1016/j.vaccine.2008.07.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Bauer L, Laksono BM, de Vrij FMS, Kushner SA, Harschnitz O, en van Riel D, ‘The neuroinvasiveness, neurotropism, and neurovirulence of SARS-CoV-2’, Trends Neurosci, vol. 45, nr. 5, pp. 358–368, mei 2022, doi: 10.1016/j.tins.2022.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Edenborough KM e.a., ‘Predicting Disease Severity and Viral Spread of H5N1 Influenza Virus in Ferrets in the Context of Natural Exposure Routes’, J Virol, vol. 90, nr. 4, pp. 1888–1897, feb. 2016, doi: 10.1128/JVI.01878-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Schrauwen EJA e.a., ‘The multibasic cleavage site in H5N1 virus is critical for systemic spread along the olfactory and hematogenous routes in ferrets’, J. Virol, vol. 86, nr. 7, pp. 3975–3984, apr. 2012, doi: 10.1128/JVI.06828-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Shinya K e.a., ‘Subclinical brain injury caused by H5N1 influenza virus infection’, J. Virol, vol. 85, nr. 10, pp. 5202–5207, mei 2011, doi: 10.1128/JVI.00239-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Iwasaki T e.a., ‘Productive infection in the murine central nervous system with avian influenza virus A (H5N1) after intranasal inoculation’, Acta Neuropathol, vol. 108, nr. 6, pp. 485–492, dec. 2004, doi: 10.1007/s00401-004-0909-0. [DOI] [PubMed] [Google Scholar]
  • [23].Park CH e.a., ‘The invasion routes of neurovirulent A/Hong Kong/483/97 (H5N1) influenza virus into the central nervous system after respiratory infection in mice’, Arch Virol, vol. 147, nr. 7, pp. 1425–1436, jul. 2002, doi: 10.1007/s00705-001-0750-x. [DOI] [PubMed] [Google Scholar]
  • [24].Tanaka H e.a., ‘Neurotropism of the 1997 Hong Kong H5N1 influenza virus in mice’, Veterinary Microbiology, vol. 95, nr. 1–2, pp. 1–13, aug. 2003, doi: 10.1016/S0378-1135(03)00132-9. [DOI] [PubMed] [Google Scholar]
  • [25].Bodewes R e.a., ‘Pathogenesis of Influenza A/H5N1 Virus Infection in Ferrets Differs between Intranasal and Intratracheal Routes of Inoculation’, The American Journal of Pathology, vol. 179, nr. 1, pp. 30–36, jul. 2011, doi: 10.1016/j.ajpath.2011.03.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Lipatov AS, Kwon YK, Pantin-Jackwood MJ, en Swayne DE, ‘Pathogenesis of H5N1 Influenza Virus Infections in Mice and Ferret Models Differs According to Respiratory Tract or Digestive System Exposure’, J Infect Dis, vol. 199, nr. 5, pp. 717–725, mrt. 2009, doi: 10.1086/596740. [DOI] [PubMed] [Google Scholar]
  • [27].Yamada M e.a., ‘Multiple routes of invasion of wild-type Clade 1 highly pathogenic avian influenza H5N1 virus into the central nervous system (CNS) after intranasal exposure in ferrets’, Acta Neuropathol, vol. 124, nr. 4, pp. 505–516, okt. 2012, doi: 10.1007/s00401-012-1010-8. [DOI] [PubMed] [Google Scholar]
  • [28].Jang H e.a., ‘Highly pathogenic H5N1 influenza virus can enter the central nervous system and induce neuroinflammation and neurodegeneration’, Proc Natl Acad Sci U S A, vol. 106, nr. 33, pp. 14063–14068, aug. 2009, doi: 10.1073/pnas.0900096106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Siegers JY e.a., ‘Vaccination Is More Effective Than Prophylactic Oseltamivir in Preventing CNS Invasion by H5N1 Virus via the Olfactory Nerve’, J Infect Dis, vol. 214, nr. 4, pp. 516–524, aug. 2016, doi: 10.1093/infdis/jiw123. [DOI] [PubMed] [Google Scholar]
  • [30].Siegers JY e.a., ‘Evolution of highly pathogenic H5N1 influenza A virus in the central nervous system of ferrets’, PLoS Pathog, vol. 19, nr. 3, p. e1011214, mrt. 2023, doi: 10.1371/journal.ppat.1011214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Plourde JR, Pyles JA, Layton RC, Vaughan SE, Tipper JL, en Harrod KS, ‘Neurovirulence of H5N1 infection in ferrets is mediated by multifocal replication in distinct permissive neuronal cell regions’, PLoS ONE, vol. 7, nr. 10, p. e46605, 2012, doi: 10.1371/journal.pone.0046605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Escada PA, Lima C, en da Silva JM, ‘The human olfactory mucosa’, Eur Arch Otorhinolaryngol, vol. 266, nr. 11, pp. 1675–1680, nov. 2009, doi: 10.1007/s00405-009-1073-x. [DOI] [PubMed] [Google Scholar]
  • [33].Jafek BW, Murrow B, Michaels R, Restrepo D, en Linschoten M, ‘Biopsies of human olfactory epithelium’, Chem Senses, vol. 27, nr. 7, pp. 623–628, sep. 2002, doi: 10.1093/chemse/27.7.623. [DOI] [PubMed] [Google Scholar]
  • [34].Gross EA, Swenberg JA, Fields S, en Popp JA, ‘Comparative morphometry of the nasal cavity in rats and mice’, J Anat, vol. 135, nr. Pt 1, pp. 83–88, aug. 1982. [PMC free article] [PubMed] [Google Scholar]
  • [35].Nibu K e.a., ‘Olfactory neuron-specific expression of NeuroD in mouse and human nasal mucosa’, Cell Tissue Res, vol. 298, nr. 3, pp. 405–414, dec. 1999, doi: 10.1007/s004419900098. [DOI] [PubMed] [Google Scholar]
  • [36].Rawson NE en Gomez G, ‘Cell and molecular biology of human olfaction’, Microsc Res Tech, vol. 58, nr. 3, pp. 142–151, aug. 2002, doi: 10.1002/jemt.10132. [DOI] [PubMed] [Google Scholar]
  • [37].de Wit E e.a., ‘1918 H1N1 Influenza Virus Replicates and Induces Proinflammatory Cytokine Responses in Extrarespiratory Tissues of Ferrets’, J Infect Dis, vol. 217, nr. 8, pp. 1237–1246, mrt. 2018, doi: 10.1093/infdis/jiy003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Shinya K e.a., ‘Avian influenza virus intranasally inoculated infects the central nervous system of mice through the general visceral afferent nerve’, Arch Virol, vol. 145, nr. 1, pp. 187–195, 2000, doi: 10.1007/s007050050016. [DOI] [PubMed] [Google Scholar]
  • [39].Plourde JR, Pyles JA, Layton RC, Vaughan SE, Tipper JL, en Harrod KS, ‘Neurovirulence of H5N1 infection in ferrets is mediated by multifocal replication in distinct permissive neuronal cell regions’, PLoS One, vol. 7, nr. 10, p. e46605, 2012, doi: 10.1371/journal.pone.0046605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Matsuda K e.a., ‘The vagus nerve is one route of transneural invasion for intranasally inoculated influenza a virus in mice’, Vet Pathol, vol. 41, nr. 2, pp. 101–107, mrt. 2004, doi: 10.1354/vp.41-2-101. [DOI] [PubMed] [Google Scholar]
  • [41].Wang X, Zhao J, Tang S, Ye Z, en Hewlett I, ‘Viremia Associated with Fatal Outcomes in Ferrets Infected with Avian H5N1 Influenza Virus’, PLoS One, vol. 5, nr. 8, p. e12099, aug. 2010, doi: 10.1371/journal.pone.0012099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Chutinimitkul S e.a., ‘H5N1 Influenza A Virus and Infected Human Plasma’, Emerging Infectious Diseases, vol. 12, nr. 6, p. 1041, jun. 2006, doi: 10.3201/eid1206.060227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].de Jong MD e.a., ‘Fatal outcome of human influenza A (H5N1) is associated with high viral load and hypercytokinemia’, Nat Med, vol. 12, nr. 10, pp. 1203–1207, okt. 2006, doi: 10.1038/nm1477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Wang X, Tan J, Zhao J, Ye Z, en Hewlett I, ‘Highly pathogenic avian influenza A virus (H5N1) can be transmitted in ferrets by transfusion’, BMC Infectious Diseases, vol. 14, nr. 1, p. 192, apr. 2014, doi: 10.1186/1471-2334-14-192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Sobata R e.a., ‘No viremia of pandemic (H1N1) 2009 was demonstrated in blood donors who had donated blood during the probable incubation period’, Transfusion, vol. 51, nr. 9, pp. 1949–1956, sep. 2011, doi: 10.1111/j.1537-2995.2011.03109.x. [DOI] [PubMed] [Google Scholar]
  • [46].Stramer SL e.a., ‘Sensitive Detection Assays for Influenza RNA Do Not Reveal Viremia in US Blood Donors’, J Infect Dis, vol. 205, nr. 6, pp. 886–894, mrt. 2012, doi: 10.1093/infdis/jir863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Reperant LA e.a., ‘Marked endotheliotropism of highly pathogenic avian influenza virus H5N1 following intestinal inoculation in cats’, J Virol, vol. 86, nr. 2, pp. 1158–1165, jan. 2012, doi: 10.1128/JVI.06375-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Alkie TN e.a., ‘Characterization of neurotropic HPAI H5N1 viruses with novel genome constellations and mammalian adaptive mutations in free-living mesocarnivores in Canada’, Emerg Microbes Infect, vol. 12, nr. 1, p. 2186608, dec. 2023, doi: 10.1080/22221751.2023.2186608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].Kumlin U, Olofsson S, Dimock K, en Arnberg N, ‘Sialic acid tissue distribution and influenza virus tropism’, Influenza Other Respir Viruses, vol. 2, nr. 5, pp. 147–154, sep. 2008, doi: 10.1111/j.1750-2659.2008.00051.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].van Riel D e.a., ‘H5N1 Virus Attachment to Lower Respiratory Tract’, Science, vol. 312, nr. 5772, p. 399, apr. 2006, doi: 10.1126/science.1125548. [DOI] [PubMed] [Google Scholar]
  • [51].van Riel D e.a., ‘Human and avian influenza viruses target different cells in the lower respiratory tract of humans and other mammals’, Am J Pathol, vol. 171, nr. 4, pp. 1215–1223, okt. 2007, doi: 10.2353/ajpath.2007.070248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Shinya K, Ebina M, Yamada S, Ono M, Kasai N, en Kawaoka Y, ‘Avian flu: influenza virus receptors in the human airway’, Nature, vol. 440, nr. 7083, pp. 435–436, mrt. 2006, doi: 10.1038/440435a. [DOI] [PubMed] [Google Scholar]
  • [53].Siegers JY e.a., ‘Viral Factors Important for Efficient Replication of Influenza A Viruses in Cells of the Central Nervous System’, J Virol, vol. 93, nr. 11, pp. e02273–18, jun. 2019, doi: 10.1128/JVI.02273-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Lin X e.a., ‘Insights into Human Astrocyte Response to H5N1 Infection by Microarray Analysis’, Viruses, vol. 7, nr. 5, pp. 2618–2640, mei 2015, doi: 10.3390/v7052618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Shinya K, Hamm S, Hatta M, Ito H, Ito T, en Kawaoka Y, ‘PB2 amino acid at position 627 affects replicative efficiency, but not cell tropism, of Hong Kong H5N1 influenza A viruses in mice’, Virology, vol. 320, nr. 2, pp. 258–266, mrt. 2004, doi: 10.1016/j.virol.2003.11.030. [DOI] [PubMed] [Google Scholar]
  • [56].Wang G e.a., ‘Apoptosis and proinflammatory cytokine responses of primary mouse microglia and astrocytes induced by human H1N1 and avian H5N1 influenza viruses’, Cell Mol Immunol, vol. 5, nr. 2, pp. 113–120, apr. 2008, doi: 10.1038/cmi.2008.14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Bauer L e.a., ‘Replication Kinetics, Cell Tropism, and Associated Immune Responses in SARS-CoV-2- and H5N1 Virus-Infected Human Induced Pluripotent Stem Cell-Derived Neural Models’, mSphere, vol. 6, nr. 3, p. e0027021, jun. 2021, doi: 10.1128/mSphere.00270-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Li Y e.a., ‘Distribution of sialic acid receptors and experimental infections with different subtypes of influenza A viruses in Qinghai-Tibet plateau wild pika’, Virol J, vol. 12, p. 63, apr. 2015, doi: 10.1186/s12985-015-0290-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Gao P e.a., ‘Biological heterogeneity, including systemic replication in mice, of H5N1 influenza A virus isolates from humans in Hong Kong’, J Virol, vol. 73, nr. 4, pp. 3184–3189, apr. 1999, doi: 10.1128/JVI.73.4.3184-3189.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Nishimura H, Itamura S, Iwasaki T, Kurata T, en Tashiro M, ‘Characterization of human influenza A (H5N1) virus infection in mice: neuro-, pneumo- and adipotropic infection’, J Gen Virol, vol. 81, nr. Pt 10, pp. 2503–2510, okt. 2000, doi: 10.1099/0022-1317-81-10-2503. [DOI] [PubMed] [Google Scholar]
  • [61].Bordes L e.a., ‘Highly Pathogenic Avian Influenza H5N1 Virus Infections in Wild Red Foxes (Vulpes vulpes) Show Neurotropism and Adaptive Virus Mutations’, Microbiol Spectr, p. e0286722, jan. 2023, doi: 10.1128/spectrum.02867-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Rimmelzwaan GF e.a., ‘Influenza A virus (H5N1) infection in cats causes systemic disease with potential novel routes of virus spread within and between hosts’, Am J Pathol, vol. 168, nr. 1, pp. 176–183; quiz 364, jan. 2006, doi: 10.2353/ajpath.2006.050466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Peng B-H e.a., ‘Neuropathology of H5N1 virus infection in ferrets’, Vet Microbiol, vol. 156, nr. 3–4, pp. 294–304, mei 2012, doi: 10.1016/j.vetmic.2011.11.025. [DOI] [PubMed] [Google Scholar]
  • [64].Kombiah S e.a., ‘Experimental pathology of two highly pathogenic H5N1 viruses isolated from crows in BALB/c mice’, Microb Pathog, vol. 141, p. 103984, apr. 2020, doi: 10.1016/j.micpath.2020.103984. [DOI] [PubMed] [Google Scholar]
  • [65].Lu X, Tumpey TM, Morken T, Zaki SR, Cox NJ, en Katz JM, ‘A mouse model for the evaluation of pathogenesis and immunity to influenza A (H5N1) viruses isolated from humans’, J Virol, vol. 73, nr. 7, pp. 5903–5911, jul. 1999, doi: 10.1128/JVI.73.7.5903-5911.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Bissel SJ, Giles BM, Wang G, Olevian DC, Ross TM, en Wiley CA, ‘Acute murine H5N1 influenza A encephalitis’, Brain Pathol, vol. 22, nr. 2, pp. 150–158, mrt. 2012, doi: 10.1111/j.1750-3639.2011.00514.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Reperant LA e.a., ‘Highly Pathogenic Avian Influenza Virus (H5N1) Infection in Red Foxes Fed Infected Bird Carcasses’, Emerg Infect Dis, vol. 14, nr. 12, pp. 1835–1841, dec. 2008, doi: 10.3201/eid1412.080470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Songserm T e.a., ‘Avian influenza H5N1 in naturally infected domestic cat’, Emerg Infect Dis, vol. 12, nr. 4, pp. 681–683, apr. 2006, doi: 10.3201/eid1204.051396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Maines TR e.a., ‘Avian Influenza (H5N1) Viruses Isolated from Humans in Asia in 2004 Exhibit Increased Virulence in Mammals’, J Virol, vol. 79, nr. 18, pp. 11788–11800, sep. 2005, doi: 10.1128/JVI.79.18.11788-11800.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Yen H-L e.a., ‘Inefficient Transmission of H5N1 Influenza Viruses in a Ferret Contact Model’, J Virol, vol. 81, nr. 13, pp. 6890–6898, jul. 2007, doi: 10.1128/JVI.00170-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Klopfleisch R e.a., ‘Encephalitis in a stone marten (Martes foina) after natural infection with highly pathogenic avian influenza virus subtype H5N1’, J Comp Pathol, vol. 137, nr. 2–3, pp. 155–159, 2007, doi: 10.1016/j.jcpa.2007.06.001. [DOI] [PubMed] [Google Scholar]
  • [72].Floyd T e.a., ‘Encephalitis and Death in Wild Mammals at a Rehabilitation Center after Infection with Highly Pathogenic Avian Influenza A(H5N8) Virus, United Kingdom’, Emerg Infect Dis, vol. 27, nr. 11, pp. 2856–2863, nov. 2021, doi: 10.3201/eid2711.211225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Thorsson E, Zohari S, Roos A, Banihashem F, Bröjer C, en Neimanis A, ‘Highly Pathogenic Avian Influenza A(H5N1) Virus in a Harbor Porpoise, Sweden’, Emerg Infect Dis, vol. 29, nr. 4, pp. 852–855, apr. 2023, doi: 10.3201/eid2904.221426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Edenborough KM e.a., ‘Predicting Disease Severity and Viral Spread of H5N1 Influenza Virus in Ferrets in the Context of Natural Exposure Routes’, J Virol, vol. 90, nr. 4, pp. 1888–1897, feb. 2016, doi: 10.1128/JVI.01878-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Lee K e.a., ‘Highly Pathogenic Avian Influenza A(H5N6) in Domestic Cats, South Korea’, Emerg Infect Dis, vol. 24, nr. 12, pp. 2343–2347, dec. 2018, doi: 10.3201/eid2412.180290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].Shinya K e.a., ‘Systemic dissemination of H5N1 influenza A viruses in ferrets and hamsters after direct intragastric inoculation’, J Virol, vol. 85, nr. 10, pp. 4673–4678, mei 2011, doi: 10.1128/JVI.00148-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].Tumpey TM, Lu X, Morken T, Zaki SR, en Katz JM, ‘Depletion of lymphocytes and diminished cytokine production in mice infected with a highly virulent influenza A (H5N1) virus isolated from humans’, J Virol, vol. 74, nr. 13, pp. 6105–6116, jul. 2000, doi: 10.1128/jvi.74.13.6105-6116.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Gao R e.a., ‘A systematic molecular pathology study of a laboratory confirmed H5N1 human case’, PLoS One, vol. 5, nr. 10, p. e13315, okt. 2010, doi: 10.1371/journal.pone.0013315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Gu J e.a., ‘H5N1 infection of the respiratory tract and beyond: a molecular pathology study’, Lancet, vol. 370, nr. 9593, pp. 1137–1145, sep. 2007, doi: 10.1016/S0140-6736(07)61515-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Zhang Z e.a., ‘Systemic infection of avian influenza A virus H5N1 subtype in humans’, Hum Pathol, vol. 40, nr. 5, pp. 735–739, mei 2009, doi: 10.1016/j.humpath.2008.08.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Rijks JM e.a., ‘Highly Pathogenic Avian Influenza A(H5N1) Virus in Wild Red Foxes, the Netherlands, 2021’, Emerg Infect Dis, vol. 27, nr. 11, pp. 2960–2962, nov. 2021, doi: 10.3201/eid2711.211281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Vreman S e.a., ‘Zoonotic Mutation of Highly Pathogenic Avian Influenza H5N1 Virus Identified in the Brain of Multiple Wild Carnivore Species’, Pathogens, vol. 12, nr. 2, p. 168, jan. 2023, doi: 10.3390/pathogens12020168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Bourgognon J-M en Cavanagh J, ‘The role of cytokines in modulating learning and memory and brain plasticity’, Brain Neurosci Adv, vol. 4, p. 2398212820979802, 2020, doi: 10.1177/2398212820979802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Zipp F, Bittner S, en Schafer DP, ‘Cytokines as emerging regulators of central nervous system synapses’, Immunity, vol. 56, nr. 5, pp. 914–925, mei 2023, doi: 10.1016/j.immuni.2023.04.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Ng YP, Lee SMY, Cheung TKW, Nicholls JM, Peiris JSM, en Ip NY, ‘Avian influenza H5N1 virus induces cytopathy and proinflammatory cytokine responses in human astrocytic and neuronal cell lines’, Neuroscience, vol. 168, nr. 3, pp. 613–623, jul. 2010, doi: 10.1016/j.neuroscience.2010.04.013. [DOI] [PubMed] [Google Scholar]
  • [86].Pringproa K e.a., ‘Tropism and Induction of Cytokines in Human Embryonic-Stem Cells-Derived Neural Progenitors upon Inoculation with Highly- Pathogenic Avian H5N1 Influenza Virus’, PLoS One, vol. 10, nr. 8, p. e0135850, 2015, doi: 10.1371/journal.pone.0135850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Short KR e.a., ‘Proinflammatory Cytokine Responses in Extra-Respiratory Tissues During Severe Influenza’, J Infect Dis, vol. 216, nr. 7, pp. 829–833, okt. 2017, doi: 10.1093/infdis/jix281. [DOI] [PubMed] [Google Scholar]
  • [88].Düsedau HP e.a., ‘Influenza A Virus (H1N1) Infection Induces Microglial Activation and Temporal Dysbalance in Glutamatergic Synaptic Transmission’, mBio, vol. 12, nr. 5, p. e0177621, okt. 2021, doi: 10.1128/mBio.01776-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Horman WSJ, Kedzierska K, Rootes CL, Bean AGD, Nguyen THO, en Layton DS, ‘Ferret Interferon (IFN)-Inducible Transmembrane Proteins Are Upregulated by both IFN-α and Influenza Virus Infection’, J Virol, vol. 95, nr. 14, p. e0011121, jun. 2021, doi: 10.1128/JVI.00111-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Jang H e.a., ‘Inflammatory effects of highly pathogenic H5N1 influenza virus infection in the CNS of mice’, J Neurosci, vol. 32, nr. 5, pp. 1545–1559, feb. 2012, doi: 10.1523/JNEUROSCI.5123-11.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Wang K e.a., ‘IP-10 Promotes Blood-Brain Barrier Damage by Inducing Tumor Necrosis Factor Alpha Production in Japanese Encephalitis’, Front Immunol, vol. 9, p. 1148, 2018, doi: 10.3389/fimmu.2018.01148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].La Mantia L, D’Amico D, Rigamonti A, Mascoli N, Bussone G, en Milanese C, ‘Interferon treatment may trigger primary headaches in multiple sclerosis patients’, Mult Scler, vol. 12, nr. 4, pp. 476–480, aug. 2006, doi: 10.1191/1352458506ms1298oa. [DOI] [PubMed] [Google Scholar]
  • [93].Hosseini S e.a., ‘Long-Term Neuroinflammation Induced by Influenza A Virus Infection and the Impact on Hippocampal Neuron Morphology and Function’, J. Neurosci, vol. 38, nr. 12, pp. 3060–3080, 21 2018, doi: 10.1523/JNEUROSCI.1740-17.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Biber K e.a., ‘Interleukin-6 upregulates neuronal adenosine A1 receptors: implications for neuromodulation and neuroprotection’, Neuropsychopharmacology, vol. 33, nr. 9, pp. 2237–2250, aug. 2008, doi: 10.1038/sj.npp.1301612. [DOI] [PubMed] [Google Scholar]
  • [95].Viviani B e.a., ‘Interleukin-1beta enhances NMDA receptor-mediated intracellular calcium increase through activation of the Src family of kinases’, J Neurosci, vol. 23, nr. 25, pp. 8692–8700, sep. 2003, doi: 10.1523/JNEUROSCI.23-25-08692.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].Balosso S e.a., ‘A novel non-transcriptional pathway mediates the proconvulsive effects of interleukin-1beta’, Brain, vol. 131, nr. Pt 12, pp. 3256–3265, dec. 2008, doi: 10.1093/brain/awn271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [97].Vezzani A en Viviani B, ‘Neuromodulatory properties of inflammatory cytokines and their impact on neuronal excitability’, Neuropharmacology, vol. 96, nr. Pt A, pp. 70–82, sep. 2015, doi: 10.1016/j.neuropharm.2014.10.027. [DOI] [PubMed] [Google Scholar]
  • [98].Katsuki H, Nakai S, Hirai Y, Akaji K, Kiso Y, en Satoh M, ‘Interleukin-1 beta inhibits long-term potentiation in the CA3 region of mouse hippocampal slices’, Eur J Pharmacol, vol. 181, nr. 3, pp. 323–326, jun. 1990, doi: 10.1016/0014-2999(90)90099-r. [DOI] [PubMed] [Google Scholar]
  • [99].Bellinger FP, Madamba S, en Siggins GR, ‘Interleukin 1 beta inhibits synaptic strength and long-term potentiation in the rat CA1 hippocampus’, Brain Res, vol. 628, nr. 1–2, pp. 227–234, nov. 1993, doi: 10.1016/0006-8993(93)90959-q. [DOI] [PubMed] [Google Scholar]
  • [100].Prieto GA, Tong L, Smith ED, en Cotman CW, ‘TNFα and IL-1β but not IL-18 Suppresses Hippocampal Long-Term Potentiation Directly at the Synapse’, Neurochem Res, vol. 44, nr. 1, pp. 49–60, jan. 2019, doi: 10.1007/s11064-018-2517-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Feinberg PA e.a., ‘Elevated TNF-α Leads to Neural Circuit Instability in the Absence of Interferon Regulatory Factor 8’, J Neurosci, vol. 42, nr. 32, pp. 6171–6185, aug. 2022, doi: 10.1523/JNEUROSCI.0601-22.2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [102].Vezzani A e.a., ‘Powerful anticonvulsant action of IL-1 receptor antagonist on intracerebral injection and astrocytic overexpression in mice’, Proc Natl Acad Sci U S A, vol. 97, nr. 21, pp. 11534–11539, okt. 2000, doi: 10.1073/pnas.190206797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Vezzani A e.a., ‘Functional role of inflammatory cytokines and antiinflammatory molecules in seizures and epileptogenesis’, Epilepsia, vol. 43 Suppl 5, pp. 30–35, 2002, doi: 10.1046/j.1528-1157.43.s.5.14.x. [DOI] [PubMed] [Google Scholar]
  • [104].Avital A e.a., ‘Impaired interleukin-1 signaling is associated with deficits in hippocampal memory processes and neural plasticity’, Hippocampus, vol. 13, nr. 7, pp. 826–834, 2003, doi: 10.1002/hipo.10135. [DOI] [PubMed] [Google Scholar]
  • [105].Zhang H e.a., ‘The distinct binding properties between avian/human influenza A virus NS1 and Postsynaptic density protein-95 (PSD-95), and inhibition of nitric oxide production’, Virol J, vol. 8, p. 298, jun. 2011, doi: 10.1186/1743-422X-8-298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Jang H e.a., ‘Highly pathogenic H5N1 influenza virus can enter the central nervous system and induce neuroinflammation and neurodegeneration’, Proc. Natl. Acad. Sci. U.S.A, vol. 106, nr. 33, pp. 14063–14068, aug. 2009, doi: 10.1073/pnas.0900096106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [107].van Riel D e.a., ‘Evidence for influenza virus CNS invasion along the olfactory route in an immunocompromised infant’, J. Infect. Dis, vol. 210, nr. 3, pp. 419–423, aug. 2014, doi: 10.1093/infdis/jiu097. [DOI] [PubMed] [Google Scholar]
  • [108].Aoki K, Nakahara Y, Yamada S, en Eto K, ‘Role of polysialic acid on outgrowth of rat olfactory receptor neurons’, Mech Dev, vol. 85, nr. 1–2, pp. 103–110, jul. 1999, doi: 10.1016/s0925-4773(99)00092-1. [DOI] [PubMed] [Google Scholar]
  • [109].de Graaf M en Fouchier RAM, ‘Role of receptor binding specificity in influenza A virus transmission and pathogenesis’, EMBO J, vol. 33, nr. 8, pp. 823–841, apr. 2014, doi: 10.1002/embj.201387442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].Schrauwen EJA, de Graaf M, Herfst S, Rimmelzwaan GF, Osterhaus ADME, en Fouchier R. a. M., ‘Determinants of virulence of influenza A virus’, Eur J Clin Microbiol Infect Dis, vol. 33, nr. 4, pp. 479–490, apr. 2014, doi: 10.1007/s10096-013-1984-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [111].Garten W e.a., ‘Influenza virus activating host proteases: Identification, localization and inhibitors as potential therapeutics’, Eur J Cell Biol, vol. 94, nr. 7–9, pp. 375–383, 2015, doi: 10.1016/j.ejcb.2015.05.013. [DOI] [PubMed] [Google Scholar]
  • [112].Böttcher-Friebertshäuser E, Klenk H-D, en Garten W, ‘Activation of influenza viruses by proteases from host cells and bacteria in the human airway epithelium’, Pathog Dis, vol. 69, nr. 2, pp. 87–100, nov. 2013, doi: 10.1111/2049-632X.12053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [113].de Bruin ACM, Funk M, Spronken MI, Gultyaev AP, Fouchier RAM, en Richard M, ‘Hemagglutinin Subtype Specificity and Mechanisms of Highly Pathogenic Avian Influenza Virus Genesis’, Viruses, vol. 14, nr. 7, p. 1566, jul. 2022, doi: 10.3390/v14071566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Schrauwen EJA e.a., ‘Insertion of a multibasic cleavage site in the haemagglutinin of human influenza H3N2 virus does not increase pathogenicity in ferrets’, J Gen Virol, vol. 92, nr. Pt 6, pp. 1410–1415, jun. 2011, doi: 10.1099/vir.0.030379-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [115].Belser JA e.a., ‘Pathogenesis of avian influenza (H7) virus infection in mice and ferrets: enhanced virulence of Eurasian H7N7 viruses isolated from humans’, J Virol, vol. 81, nr. 20, pp. 11139–11147, okt. 2007, doi: 10.1128/JVI.01235-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [116].Taft AS e.a., ‘Identification of mammalian-adapting mutations in the polymerase complex of an avian H5N1 influenza virus’, Nat Commun, vol. 6, p. 7491, jun. 2015, doi: 10.1038/ncomms8491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [117].Yu Z, Cheng K, Sun W, Zhang X, Xia X, en Gao Y, ‘PB2 and HA mutations increase the virulence of highly pathogenic H5N5 clade 2.3.4.4 avian influenza virus in mice’, Arch Virol, vol. 163, nr. 2, pp. 401–410, feb. 2018, doi: 10.1007/s00705-017-3631-7. [DOI] [PubMed] [Google Scholar]
  • [118].Schmolke M e.a., ‘Differential contribution of PB1-F2 to the virulence of highly pathogenic H5N1 influenza A virus in mammalian and avian species’, PLoS Pathog, vol. 7, nr. 8, p. e1002186, aug. 2011, doi: 10.1371/journal.ppat.1002186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [119].Varga ZT e.a., ‘The influenza virus protein PB1-F2 inhibits the induction of type I interferon at the level of the MAVS adaptor protein’, PLoS Pathog, vol. 7, nr. 6, p. e1002067, jun. 2011, doi: 10.1371/journal.ppat.1002067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [120].Ji Z-X, Wang X-Q, en Liu X-F, ‘NS1: A Key Protein in the “Game” Between Influenza A Virus and Host in Innate Immunity’, Front Cell Infect Microbiol, vol. 11, p. 670177, 2021, doi: 10.3389/fcimb.2021.670177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [121].Spesock A e.a., ‘The virulence of 1997 H5N1 influenza viruses in the mouse model is increased by correcting a defect in their NS1 proteins’, J Virol, vol. 85, nr. 14, pp. 7048–7058, jul. 2011, doi: 10.1128/JVI.00417-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [122].Twu KY, Kuo R-L, Marklund J, en Krug RM, ‘The H5N1 influenza virus NS genes selected after 1998 enhance virus replication in mammalian cells’, J Virol, vol. 81, nr. 15, pp. 8112–8121, aug. 2007, doi: 10.1128/JVI.00006-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [123].Brask J, Chauhan A, Hill RH, Ljunggren H-G, en Kristensson K, ‘Effects on synaptic activity in cultured hippocampal neurons by influenza A viral proteins’, J Neurovirol, vol. 11, nr. 4, pp. 395–402, aug. 2005, doi: 10.1080/13550280500186916. [DOI] [PubMed] [Google Scholar]
  • [124].Govorkova EA, Ilyushina NA, Boltz DA, Douglas A, Yilmaz N, en Webster RG, ‘Efficacy of oseltamivir therapy in ferrets inoculated with different clades of H5N1 influenza virus’, Antimicrob Agents Chemother, vol. 51, nr. 4, pp. 1414–1424, apr. 2007, doi: 10.1128/AAC.01312-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [125].Boltz DA, Rehg JE, McClaren J, Webster RG, en Govorkova EA, ‘Oseltamivir prophylactic regimens prevent H5N1 influenza morbidity and mortality in a ferret model’, J Infect Dis, vol. 197, nr. 9, pp. 1315–1323, mei 2008, doi: 10.1086/586711. [DOI] [PubMed] [Google Scholar]
  • [126].Taniguchi K e.a., ‘Characterization of the In Vitro and In Vivo Efficacy of Baloxavir Marboxil against H5 Highly Pathogenic Avian Influenza Virus Infection’, Viruses, vol. 14, nr. 1, p. 111, jan. 2022, doi: 10.3390/v14010111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [127].Morimoto K e.a., ‘Oseltamivir (Tamiflu) efflux transport at the blood-brain barrier via P-glycoprotein’, Drug Metab Dispos, vol. 36, nr. 1, pp. 6–9, jan. 2008, doi: 10.1124/dmd.107.017699. [DOI] [PubMed] [Google Scholar]
  • [128].Ose A e.a., ‘P-glycoprotein restricts the penetration of oseltamivir across the blood-brain barrier’, Drug Metab Dispos, vol. 36, nr. 2, pp. 427–434, feb. 2008, doi: 10.1124/dmd.107.018556. [DOI] [PubMed] [Google Scholar]
  • [129].Chestakova Irina V. e.a., ‘High number of HPAI H5 Virus Infections and Antibodies in Wild Carnivores in the Netherlands, 2020–2022’, bioRxiv, p. 2023.05.12.540493, jan. 2023, doi: 10.1101/2023.05.12.540493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [130].Fulton CDM, Beasley DWC, Bente DA, en Dineley KT, ‘Long-term, West Nile virus-induced neurological changes: A comparison of patients and rodent models’, Brain Behav Immun Health, vol. 7, p. 100105, aug. 2020, doi: 10.1016/j.bbih.2020.100105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Kim M e.a., ‘Comparative analyses of influenza virus receptor distribution in the human and mouse brains’, J Chem Neuroanat, vol. 52, pp. 49–57, sep. 2013, doi: 10.1016/j.jchemneu.2013.05.002. [DOI] [PubMed] [Google Scholar]
  • [132].Ning Z-Y, Luo M-Y, Qi W-B, Yu B, Jiao P-R, en Liao M, ‘Detection of expression of influenza virus receptors in tissues of BALB/c mice by histochemistry’, Vet Res Commun, vol. 33, nr. 8, pp. 895–903, dec. 2009, doi: 10.1007/s11259-009-9307-3. [DOI] [PubMed] [Google Scholar]
  • [133].Takahashi M, Yamada T, Nakajima S, Nakajima K, Yamamoto T, en Okada H, ‘The substantia nigra is a major target for neurovirulent influenza A virus’, J Exp Med, vol. 181, nr. 6, pp. 2161–2169, jun. 1995, doi: 10.1084/jem.181.6.2161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [134].Yao L, Korteweg C, Hsueh W, en Gu J, ‘Avian influenza receptor expression in H5N1-infected and noninfected human tissues’, FASEB J, vol. 22, nr. 3, pp. 733–740, mrt. 2008, doi: 10.1096/fj.06-7880com. [DOI] [PubMed] [Google Scholar]
  • [135].Nelli RK, Kuchipudi SV, White GA, Perez BB, Dunham SP, en Chang K-C, ‘Comparative distribution of human and avian type sialic acid influenza receptors in the pig’, BMC Vet Res, vol. 6, p. 4, jan. 2010, doi: 10.1186/1746-6148-6-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [136].Wang H, Wu X, Cheng Y, An Y, en Ning Z, ‘Tissue distribution of human and avian type sialic acid influenza virus receptors in domestic cat’, Acta Vet Hung, vol. 61, nr. 4, pp. 537–546, dec. 2013, doi: 10.1556/AVet.2013.030. [DOI] [PubMed] [Google Scholar]
  • [137].Ning Z-Y e.a., ‘Tissue distribution of sialic acid-linked influenza virus receptors in beagle dogs’, J Vet Sci, vol. 13, nr. 3, pp. 219–222, sep. 2012, doi: 10.4142/jvs.2012.13.3.219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [138].Nicholls JM, Bourne AJ, Chen H, Guan Y, en Peiris JM, ‘Sialic acid receptor detection in the human respiratory tract: evidence for widespread distribution of potential binding sites for human and avian influenza viruses’, Respir Res, vol. 8, nr. 1, p. 73, 2007, doi: 10.1186/1465-9921-8-73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [139].Rimmelzwaan GF, Kuiken T, van Amerongen G, Bestebroer TM, Fouchier RA, en Osterhaus AD, ‘Pathogenesis of influenza A (H5N1) virus infection in a primate model’, J Virol, vol. 75, nr. 14, pp. 6687–6691, jul. 2001, doi: 10.1128/JVI.75.14.6687-6691.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [140].Rowe T, Cho DS, Bright RA, Zitzow LA, en Katz JM, ‘Neurological Manifestations of Avian Influenza Viruses in Mammals’, avdi, vol. 47, nr. s3, pp. 1122–1126, sep. 2003, doi: 10.1637/0005-2086-47.s3.1122. [DOI] [PubMed] [Google Scholar]
  • [141].Zitzow LA, Rowe T, Morken T, Shieh W-J, Zaki S, en Katz JM, ‘Pathogenesis of avian influenza A (H5N1) viruses in ferrets’, J Virol, vol. 76, nr. 9, pp. 4420–4429, mei 2002, doi: 10.1128/jvi.76.9.4420-4429.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [142].Lipatov AS e.a., ‘Neurovirulence in mice of H5N1 influenza virus genotypes isolated from Hong Kong poultry in 2001’, J Virol, vol. 77, nr. 6, pp. 3816–3823, mrt. 2003, doi: 10.1128/jvi.77.6.3816-3823.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [143].Sun R, Luo J, Gao Y, en He H, ‘Different infection routes of avian influenza A (H5N1) virus in mice’, Integr Zool, vol. 4, nr. 4, pp. 402–408, dec. 2009, doi: 10.1111/j.1749-4877.2009.00178.x. [DOI] [PubMed] [Google Scholar]
  • [144].Shestopalov AM e.a., ‘Studying the pathogenicity of avian influenza virus subtype H5N1 strains from the Russian Federation using mouse model’, Bull Exp Biol Med, vol. 146, nr. 3, pp. 341–343, sep. 2008, doi: 10.1007/s10517-008-0296-y. [DOI] [PubMed] [Google Scholar]
  • [145].Kim HM, Park EH, Yum J, Kim HS, en Seo SH, ‘Greater virulence of highly pathogenic H5N1 influenza virus in cats than in dogs’, Arch Virol, vol. 160, nr. 1, pp. 305–313, jan. 2015, doi: 10.1007/s00705-014-2284-z. [DOI] [PubMed] [Google Scholar]
  • [146].Herfst S e.a., ‘A Dutch highly pathogenic H5N6 avian influenza virus showed remarkable tropism for extra-respiratory organs and caused severe disease but was not transmissible via air in the ferret model’, mSphere, p. e0020023, jul. 2023, doi: 10.1128/msphere.00200-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [147].Songserm T e.a., ‘Fatal avian influenza A H5N1 in a dog’, Emerg Infect Dis, vol. 12, nr. 11, pp. 1744–1747, nov. 2006, doi: 10.3201/eid1211.060542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [148].Roberton SI e.a., ‘Avian influenza H5N1 in viverrids: implications for wildlife health and conservation’, Proc Biol Sci, vol. 273, nr. 1595, pp. 1729–1732, jul. 2006, doi: 10.1098/rspb.2006.3549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [149].Weber S e.a., ‘Molecular analysis of highly pathogenic avian influenza virus of subtype H5N1 isolated from wild birds and mammals in northern Germany’, J Gen Virol, vol. 88, nr. Pt 2, pp. 554–558, feb. 2007, doi: 10.1099/vir.0.82300-0. [DOI] [PubMed] [Google Scholar]
  • [150].He S, Shi J, Qi X, Huang G, Chen H, en Lu C, ‘Lethal infection by a novel reassortant H5N1 avian influenza A virus in a zoo-housed tiger’, Microbes Infect, vol. 17, nr. 1, pp. 54–61, jan. 2015, doi: 10.1016/j.micinf.2014.10.004. [DOI] [PubMed] [Google Scholar]
  • [151].Hu T e.a., ‘Fatal influenza A (H5N1) virus Infection in zoo-housed Tigers in Yunnan Province, China’, Sci Rep, vol. 6, p. 25845, mei 2016, doi: 10.1038/srep25845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [152].Postel A e.a., ‘Infections with highly pathogenic avian influenza A virus (HPAIV) H5N8 in harbor seals at the German North Sea coast, 2021’, Emerg Microbes Infect, vol. 11, nr. 1, pp. 725–729, dec. 2022, doi: 10.1080/22221751.2022.2043726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [153].Tammiranta N e.a., ‘Highly pathogenic avian influenza A (H5N1) virus infections in wild carnivores connected to mass mortalities of pheasants in Finland’, Infect Genet Evol, p. 105423, mrt. 2023, doi: 10.1016/j.meegid.2023.105423. [DOI] [PubMed] [Google Scholar]
  • [154].Puryear W e.a., ‘Outbreak of Highly Pathogenic Avian Influenza H5N1 in New England Seals’. bioRxiv, p. 2022.07.29.501155, 30 juli 2022. doi: 10.1101/2022.07.29.501155. [DOI] [Google Scholar]
  • [155].Leguia M e.a., ‘Highly pathogenic avian influenza A (H5N1) in marine mammals and seabirds in Peru’. bioRxiv, p. 2023.03.03.531008, 3 maart 2023. doi: 10.1101/2023.03.03.531008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [156].Murawski A e.a., ‘Highly pathogenic avian influenza A(H5N1) virus in a common bottlenose dolphin (Tursiops truncatus) in Florida’, ResearchSquare, 17 juli 2023. Accessed 27 July 2023. doi: https://www.researchsquare.com/article/rs-3065313/v1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [157].Yuen KY e.a., ‘Clinical features and rapid viral diagnosis of human disease associated with avian influenza A H5N1 virus’, Lancet, vol. 351, nr. 9101, pp. 467–471, feb. 1998, doi: 10.1016/s0140-6736(98)01182-9. [DOI] [PubMed] [Google Scholar]
  • [158].To KF e.a., ‘Pathology of fatal human infection associated with avian influenza A H5N1 virus’, J Med Virol, vol. 63, nr. 3, pp. 242–246, mrt. 2001, doi: . [DOI] [PubMed] [Google Scholar]
  • [159].Uiprasertkul M e.a., ‘Influenza A H5N1 replication sites in humans’, Emerg Infect Dis, vol. 11, nr. 7, pp. 1036–1041, jul. 2005, doi: 10.3201/eid1107.041313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [160].Chokephaibulkit K e.a., ‘A child with avian influenza A (H5N1) infection’, Pediatr Infect Dis J, vol. 24, nr. 2, pp. 162–166, feb. 2005, doi: 10.1097/01.inf.0000151037.25237.1e. [DOI] [PubMed] [Google Scholar]
  • [161].de Jong MD e.a., ‘Fatal avian influenza A (H5N1) in a child presenting with diarrhea followed by coma’, N Engl J Med, vol. 352, nr. 7, pp. 686–691, feb. 2005, doi: 10.1056/NEJMoa044307. [DOI] [PubMed] [Google Scholar]
  • [162].Writing Committee of the Second World Health Organization Consultation on Clinical Aspects of Human Infection with Avian Influenza A (H5N1) Virus e.a., ‘Update on avian influenza A (H5N1) virus infection in humans’, N Engl J Med, vol. 358, nr. 3, pp. 261–273, jan. 2008, doi: 10.1056/NEJMra0707279. [DOI] [PubMed] [Google Scholar]
  • [163].Kandun IN e.a., ‘Three Indonesian clusters of H5N1 virus infection in 2005’, N Engl J Med, vol. 355, nr. 21, pp. 2186–2194, nov. 2006, doi: 10.1056/NEJMoa060930. [DOI] [PubMed] [Google Scholar]
  • [164].Beigel JH e.a., ‘Avian influenza A (H5N1) infection in humans’, N Engl J Med, vol. 353, nr. 13, pp. 1374–1385, sep. 2005, doi: 10.1056/NEJMra052211. [DOI] [PubMed] [Google Scholar]
  • [165].Yu H e.a., ‘Clinical Characteristics of 26 Human Cases of Highly Pathogenic Avian Influenza A (H5N1) Virus Infection in China’, PLoS One, vol. 3, nr. 8, p. e2985, aug. 2008, doi: 10.1371/journal.pone.0002985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [166].Oner AF e.a., ‘Avian influenza A (H5N1) infection in eastern Turkey in 2006’, N Engl J Med, vol. 355, nr. 21, pp. 2179–2185, nov. 2006, doi: 10.1056/NEJMoa060601. [DOI] [PubMed] [Google Scholar]
  • [167].Mak GCK, Kwan MY-W, Mok CKP, Lo JYC, Peiris M, en Leung CW, ‘Influenza A(H5N1) Virus Infection in a Child With Encephalitis Complicated by Obstructive Hydrocephalus’, Clin Infect Dis, vol. 66, nr. 1, pp. 136–139, jan. 2018, doi: 10.1093/cid/cix707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [168].Rajabali N, Lim T, Sokolowski C, Prevost JD, en Lee EZ, ‘Avian influenza A (H5N1) infection with respiratory failure and meningoencephalitis in a Canadian traveller’, Can J Infect Dis Med Microbiol, vol. 26, nr. 4, pp. 221–223, aug. 2015, doi: 10.1155/2015/961080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [169].Zhang L e.a., ‘Clinical features of the first critical case of acute encephalitis caused by the avian influenza A (H5N6) virus’, Emerg Microbes Infect, vol. 11, nr. 1, pp. 2437–2446, dec. 2022, doi: 10.1080/22221751.2022.2122584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [170].Anis A, AboElkhair M, en M Ibrahim, ‘Characterization of highly pathogenic avian influenza H5N8 virus from Egyptian domestic waterfowl in 2017’, Avian Pathol, vol. 47, nr. 4, pp. 400–409, aug. 2018, doi: 10.1080/03079457.2018.1470606. [DOI] [PubMed] [Google Scholar]
  • [171].Bányai K e.a., ‘Neuroinvasive influenza virus A(H5N8) in fattening ducks, Hungary, 2015’, Infect Genet Evol, vol. 43, pp. 418–423, sep. 2016, doi: 10.1016/j.meegid.2016.05.027. [DOI] [PubMed] [Google Scholar]
  • [172].van den Brand JM e.a., ‘Host-specific exposure and fatal neurologic disease in wild raptors from highly pathogenic avian influenza virus H5N1 during the 2006 outbreak in Germany’, Vet Res, vol. 46, p. 24, mrt. 2015, doi: 10.1186/s13567-015-0148-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [173].Caliendo V, Leijten L, van de Bildt MWG, Fouchier RAM, Rijks JM, en Kuiken T, ‘Pathology and virology of natural highly pathogenic avian influenza H5N8 infection in wild Common buzzards (Buteo buteo)’, Sci Rep, vol. 12, nr. 1, p. 920, jan. 2022, doi: 10.1038/s41598-022-04896-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [174].Djurdjević B e.a., ‘Highly Pathogenic Avian Influenza H5N8 Outbreak in Backyard Chickens in Serbia’, Animals (Basel), vol. 13, nr. 4, p. 700, feb. 2023, doi: 10.3390/ani13040700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [175].Fujimoto Y e.a., ‘Experimental and natural infections of white-tailed sea eagles (Haliaeetus albicilla) with high pathogenicity avian influenza virus of H5 subtype’, Front Microbiol, vol. 13, p. 1007350, 2022, doi: 10.3389/fmicb.2022.1007350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [176].Krone O e.a., ‘White-Tailed Sea Eagle (Haliaeetus albicilla) Die-Off Due to Infection with Highly Pathogenic Avian Influenza Virus, Subtype H5N8, in Germany’, Viruses, vol. 10, nr. 9, p. 478, sep. 2018, doi: 10.3390/v10090478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [177].Pantin-Jackwood MJ en Swayne DE, ‘Pathogenesis and pathobiology of avian influenza virus infection in birds’, Rev Sci Tech, vol. 28, nr. 1, pp. 113–136, apr. 2009. [PubMed] [Google Scholar]
  • [178].Pantin-Jackwood M, ‘Chapter 13. Pathobiology of avian influenza in domestic ducks.’, in Animal Influenza, 2nd dr.Wiley-Blackwell, Hoboken, New Jersey, 2016. [Google Scholar]
  • [179].Caliendo V e.a., ‘Transatlantic spread of highly pathogenic avian influenza H5N1 by wild birds from Europe to North America in 2021’, Sci Rep, vol. 12, nr. 1, p. 11729, jul. 2022, doi: 10.1038/s41598-022-13447-z. [DOI] [PMC free article] [PubMed] [Google Scholar]

Resources

RESOURCES