Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Mar 1.
Published in final edited form as: Clin Lab Med. 2010 Mar;30(1):67–91. doi: 10.1016/j.cll.2010.01.004

Hantaviruses

Mohammed Mir 1
PMCID: PMC2880890  NIHMSID: NIHMS177115  PMID: 20513542

In 1978, the etiologic agent of Korean Hemerologic fever was isolated from small infected field rodent Apodemus agrarius near Hantan river in South Korea. The virus was named as Hantaan virus, after the name of the river Hantan. This initial discovery dates back to scientific approaches that were initiated after the Korean war (1951-1953), during which more than 3000 cases of Korean hemorrhagic fever were reported among UN troops. In 1981, Hantaan virus strain 76-118, isolated from Apodemus agrarius was grown in A549 cell line, and its electron microscopic images revealed that the virus was a new member of the Bunyaviridae family. It was observed that hantaviruses unlike other members of this family do not have an arthropod vector, and exclusively establish a persistent infection in the population of their specific rodent hosts. In 1981, a new genus termed as “hantavirus” was introduced in the Bunyaviridae family, which included the viruses that cause hemoroligic fever with renal syndrome (HFRS) (Fig1). It was initially thought that pathogenic hantaviruses are restricted to old word. Until 1993, the only native hantavirus found in new word was non pathogenic Prospect Hill virus (PHV). This myth ended after the hantavirus outbreak in the four corner region of Southwestern United States that caused serious respiratory distress in infected patients and lead to the discovery of a new hantavirus disease called hantavirus cardiopulmonary syndrome (HPS). An examination of frozen stored samples of lung tissue from people who had died of unexplained lung disease in the past revealed that HPS is an old disease with conformed cases dating to at least 1959. Within a very short period the virus causing HPS was isolated from common dear mouse (Peromyscus maniculatus, Fig 1) and was later named as Sin Nombre virus (SNV). Later on, it became clear that other hantaviruses similar to SNV, such as Andes virus (ANDV), are present through out the united states. Currently, the hantavirus genus includes more than twenty one species and more than 30 genotypes (Table 1).

Figure 1. Classification of negative stranded RNA viruses and the rodent reservoirs for the hantavirus genus.

Figure 1

Left panel shows the classification of negative stranded RNA viruses in two groups, Mononegavirales and segmented negative stranded RNA viruses. Mononegavirales have a single copy of negative sense RNA genome and segmented RNA viruses have multiple copies of negative sense RNA genome. Segmented RNA viruses have been further classified into three families, Orthomyxoviridae, Bunyaviridae and Arenaviridae. Viruses in the Bunyaviridae family have been classified into five genera, Bunyavirus, Hantavirus, Nairovirus, Phlebovirus and Tospovirus. Right panel show the rodent reservoirs for hantaviruses. (A) Apodemus agrarius (Reservoir for hantaan virus that cause HFRS); (B) deer mouse (Peromyscus maniculatus), (C) The Cotton Rat (Sigmodon hispidus); (D) The Rice Rat (Oryzomys palustris); (E) The White-footed Mouse (Peromyscus leucopus). Rodents in B, C, D and E cause HPS. All pictures in the right panel were obtained from the CDC web site; http://www.cdc.gov/NCIDOD/DISEASES/HANTA/HPS/noframes/rodents.htm

Table 1.

Members of the genus Hantavirus, family Bunyaviridae

Species Disease Principal
Reservoir
Distribution
of Virus
Distribution of Reservoir
Hantaan (HTN) HFRSa Apodemus agrarius
(striped field mouse)
China,
Russia, Korea
C Europe south to Thrace, Caucasus,
& Tien Shan Mtns; Amur River
through Korea to E Xizang & E
Yunnan, W Sichuan, Fujiau, &
Taiwan(China)
Dobrava-
Belgrade
(DOB)
HFRS Apodemus flavicollis
(yellow-neck
mouse)
Balkans England & Wales, from NW Spain,
France, S Scandinavia through
European Russia to Urals, S Italy, the
Balkans, Syria, Lebanon, & Israel
Seoul (SEO) HFRS Rattus norvegicus
(Norway rat)
Worldwide Worldwide
Puumala
(PUU)
HFRS Clethrionomys
glareolus
(bank vole)
Europe, Russia,
Scandinavia
W Palearctic from France and
Scandinavia to Lake Baikai, south to
N Spain, N Italy, Balkans, W Turkey,
N Kazakhstan, Altai & Sayan Mtns;
Britain & SW Ireland
Thailand
(THAI)
ndb Bandicota indica
(bandicoot rat)
Thailand Sri Lanka, peninsular India to Nepal,
Burma, NE India, S China, Laos,
Taiwan, Thailand, Vietnam
Prospect Hill
(PH)
nd Microtus
pennsylvanicus
(meadow vole)
U.S., Canada C Alaska to Labrador, including
Newfoundland & Prince Edward
Island, Canada; Rocky Mountains to
N New Mexico, in Great Plains to N
Kansas, & in Appalachians to N
Georgia, U.S.
Khabarovsk
(KHB)
nd Microtus fortis
(reed vole)
Russia Transbaikalia Amur region; E China
Thottapalayam
(TPM)
nd Suncus murinus
(musk shrew)
India Afghanistan, Pakistan, India, Sri
Lanka, Nepal, Bhutan, Burma, China,
Taiwan, Japan, Indomalayan Region
Tula (TUL) nd Microtus arvalis
(European common
vole)
Europe Throughout Europe to Black Sea &
NE to Kirov region, Russia
Sin Nombre
(SN)
HPSc Peromyscus
maniculatus
(deer mouse)
U.S., Canada, Alaska Panhandle across N Mexico
Canada, south through most of
continental U.S., excluding SE & E
seaboard, to southernmost Baja
California Sur and to NC Oaxaca,
Mexico
New York (NY) HPS Peromyscus
leucopus
(white-footed
mouse)
U.S. C and E U.S. to S Alberta & S
Ontario, Quebec & Nova Scotia,
Canada; to N Durango & along
Caribbean coast to Isthmus of
Tehuantepec & Yucatan Peninsula,
Mexico
Black Creek
Canal
(BCC)
HPS Sigmodon hispidus
(cotton rat)
U.S. SE U.S., from S Nebraska to C
Virginia south to SE Arizona &
peninsular Florida; interior & E Mexico
through Middle America to C Panama;
in South Amer ica to N Colombia & N
Venezuela
El Moro
Canyon
(ELMC) d
nd Reithrodontomys
megalotis
(Western harvest
mouse)
U.S., Mexico British Columbia & SE Alberta,
Canada; W and NC U.S., S to N Baja
California & interior Mexico to central
Oaxaca
Bayou (BAY) d HPS Oryzomys palustris
(rice rat)
U.S. SE Kansas to E Texas, eastward to S
New Jersey & peninsular Florida
Topografov
(TOP)
nd Lemmus sibiricus
(Siberian lemming)
Siberia Palearctic, from White Sea, W Russia,
to Chukotski Peninsula, NE Siberia, &
Kamchatka; Nearctic, from W Alaska
E to Baffin Island & Hudson Bay, S
Rocky Mtns to C B.C., Canada
Andes (AND) HPS Oligoryzomys
longicaudatusf
(long-tailed pygmy
rice rat)
Argentina NC to S Andes, approximately to 50
deg S latitude, in Chile & Argentina
Isla Vista
(ISLA) d
nd Microtus californicus
(California vole)
U.S. Pacific coast, from SW Oregon
through California, U.S., to N Baja
California, Mexico
Bloodland
Lake
(BLL) d
nd Microtus
ochrogaster
(prairie vole)
U.S. N & C Great Plains, EC Alberta to S
Manitoba, Canada, S to N Oklahoma
& Arkansas, E to C Tennessee & W
West Virginia, U.S.; relic populations
elsewhere in U.S. & Mexico
Muleshoe
(MUL) d
nd Sigmodon hipidus
(cotton rat)
U.S. See Black Creek Canal
Rio Segundo
(RIOS) d
nd Reithrodontomys
mexicanus
(Mexican harvest
mouse)
Costa Rica S Tamaulipas & WC Michoacan,
Mexico, S through Middle American
highlands to W Panama; Andes of W
Colombia & N Ecuador
Rio Mamore
(RIOM) d
nd Oligoryzomys
microtis
(small-eared pygmy
rice rat)
Bolivia C Brazil south of Rios Solimoes-
Amazon & contiguous low lands of
Peru, Bolivia, Paraguay, & Argentina.
Oran virus HPS “Oligoryzomys
longicaudatus” *
(Northern Argentina
South America South America
Hu39694 HPS Oligoryzomys f.
flavescens
South America
(Argentina
South America (Argentina
Laguna Negra
virus
HPS Calomys laucha South America South America
Choclo virus HPS Oligoryzomys
fulvescens
South America South America
Juquitiba virus HPS Oligoryzomys
nigripes
South America
(Brazil
South America (Brazil
Araraquara
virus
HPS Bolomys lasiurus South America
(Brazil
South America (Brazil
Castelo Dos
Sonhos virus
HPS Oligoryzomyys ssp
f.
South America
(Brazil
South America (Brazil
Araucaria
virus
HPS Bolomys lasiurus
or Akodon ssp f.
South America
(Brazil)
South America (Brazil)

Note: The table contents are from references (14, 27)

a

HFRS, hemorrhagic fever with renal syndrome

b

nd, none documented

c

HPS, hantavirus pulmonary syndrome

d

not yet isolated in cell culture

e

viruses for which incomplete characterization is available, but for which there is clear evidence indicating that they are unique

f

suspected host, but not confirmed

Hantaviruses have coevolved for millions of years with their rodent and insectivore reservoirs. Rodent reservoirs include both Cricetidae rodents (subfamilies Arvicolinae, Neotominae and Sigmodontinae) and Muridae rodents (subfamily Murinae). Cricetidae rodents include voles, lemmings of the northern hemisphere and new world mice and rats. Muridae rodents include old world mice and rats (Fig 2). Hantavirus phylogeny follows closely that of their rodent hosts, suggesting long term co-evolution, although there are evidences of occasional host switches in the past. The phylogenetic tree suggests that co-speciation of hantavirus with their host animals of four different rodent subfamilies appears to influence their ability to cause a specific clinical manifestation in humans. For example, a majority of viruses in Neotominae and Sigmodontinae subfamilies are known to cause severe HPS with high mortality rate (40-50%). These viruses are distributed throughout North and South America in different rodent species of New World Neotominae and Sigmodontinae rodents. Old world hantaviruses that have co-evolved with Murinae rodents cause severe HFRS that primarily affects kidney function with a mortality of 0-15%. Although the disease caused by Murinae viruses has less mortality rate, it still poses a significant threat to human health due to severity of the disease and the ability of the viruses to cause large scale epidemics. Among Arvicolinae-borne hataviruses, only puumala virus (PUUV) causes a mild form of human disease often referred as nephropathi epidemia with a mortality rate of less than 1%. Interestingly, most other members of this subfamily are non-pathogenic to humans. Until recently, the only exception that did not have a confirmed rodent connection is Thottapalayam virus (TPMV), which was isolated from an Asian house shrew or musk shrew (Suncus murinus) captured in 1964 during a survey for Japanese encephalitis virus in southern India (4).

Figure 2.

Figure 2

Phylogenic tree of hantaviruses carried by the different rodents (Family Muridae, subfamily Murinae and Family Cricetidae, subfamilies Arvicolinae, Sigmodontinae and Neotominae) and insectivores. The tree is based on the complete coding region of the S segment. HTNV, Hantaan virus; SEOV, Seoul virus; DOBV, Dobrava virus; SAAV, Saaremaa virus; PUUV, Puumala virus, TULV, Tula virus; PHV, Prospect Hill virus; BLLV, Blood Land Lake virus; ISLAV, Isla Vista virus; KHAV, Khabarovsk virus; TOPV, Topografov virus; SNV, Sin Nombre virus; NYV, New York virus; MGLV, Monongahela virus; ELMCV, El Moro Canyon virus; RIOSV, Rio Segundo virus; MULV, Muleshoe virus; BAYO, Bayou virus; BCCV, Black Creek Canal virus; LANV, Laguna Negra virus; RIOMV, Rio Mamore virus; ANDV, Andes virus; TPMV, Thottapalayam virus. In the figure, viruses causing HFRS are in red type and those causing HCPS in blue type. Viruses not associated with disease are in black type. This picture was obtained from reference (33)

Microbiology

Electron microscopy reveled that hantaviruses are spherical or oval particles with a diameter of 80 to 210 nm (Fig 3). They have tripartite negative sense RNA genome (13, 28). The large (L) segment genomic RNA encodes viral RNA dependent RNA polymerase (RdR), the medium size (M) segment encodes viral glycoprotein precursor (GPC) which is later cleaved into two glycoproteins G1 and G2, and the small (S) segment encodes the viral nucleocapsid protein (N). The nucleotide sequence at 5′ and 3′ termini of each genome segment is complementary and undergoes base pairing to form panhandle structures (Fig 3). Inside the virus particle the three genomic RNAs are complexed with N protein and form three individual nucleocapsids, which along with RdRp are packaged within a lipid envelop. Two glycoproteins G1 and G2 remain embedded within the lipid envelop (Fig 3).

Figure 3. Hantaviral genome.

Figure 3

Figure 3

(a) Hantaviral genome comprises of three negative sense RNAs, S segment encodes nucleocapsid protein (N), M segment encodes glycoproteins G1 and G2, and L segment encodes viral RdRp. (b) Panhandle structures of three hantaviral genomic RNAs are formed by the base pairing of complementary bases at 5′ and 3′ terminus of each genome segment. (C) Pictorial representation of hantavirus particle, showing three nucleocapsids enveloped in a lipid bilayer. (d) Thin-section electron micrograph of Sin Nombre virus isolate, a causative agent of hantavirus pulmonary syndrome (HPS) http://www.cdc.gov/ncidod/diseases/hanta/hps/noframes/hpsem.htm. (e) Pictorial representation of hantavirus life cycle.

Like many other viruses, hantaviruses enter the host cells by the interaction between viral glycoproteins and cell surface integrin receptors (Fig 3). Interestingly, pathogenic and non pathogenic hantavirus use different integrin receptors to enter their host cells. Human integrins αIIaβ3 expressed by platelets, and αvβ3 expressed by endothelial cells mediate the cellular entry for HFRS and HCP causing hantaviruses (10). In contrast, non pathogenic hantaviruses, such as, PHV and TULV use α5β1 receptor for cellular entry. Once engaged on the host cell surface, hantaviruses enter the cells by clathrin-dependent endocytosis. Upon internalization, the three nucleocapsids are released into the cell cytoplasm along with viral RdRp. Subsequently, RdRp initiates transcription and viral mRNAs encoding three viral proteins are synthesized. Viral mRNAs are around 100 nucleotides shorter than parent genomic viral RNAs, and lack poly A tails. Viral RdRp uses a novel “cap snatching” mechanism for transcription initiation. During cap snatching 10-14 nucleotide long 5′ capped oligonucleotides are cleaved from host cell transcripts and used as primers by viral RdRp to initiate the viral mRNA synthesis, following a prime-and-realign mechanism (7, 21). Viral mRNAs are translated in the cell cytoplasm by the host cell translation machinery. Viral RdRp also replicates the genome via complementary cRNA synthesis. Complementary cRNAs are exact complement of genomic viral RNA (vRNA), and serve as templates for the synthesis of negative sense viral genome. Viral assembly and maturation takes place either on the cell surface or on the Golgi. Virions that mature on the Golgi are transported to cell surface via vesicular secretory pathways. Ultimately, new virions bud off the host cells from plasma membrane.

Nucleocapsid protein

N protein is the most abundant hantavirus protein found in the cytoplasm of infected cells. Its transcript is detected in infected cells six hours post infection. N protein is responsible for encapsidation and packaging of the viral genome. However, recent studies have shown that N is a multifunctional protein involved in diverse viral functions, including its role in the transcription and translation initiation of viral mRNA (21, 22). Trimeric N also recognizes vRNA panhandle with specificity and likely facilitates the selective incorporation of viral genomic RNA into virions. N has also been found to interact with multiple host cell proteins and the nature of such interaction remains unclear.

Glycoproteins

Glycoprotein precursor (GPC) is synthesized on ribosomes associated with endoplasmic reticulum (ER) and is translocated to ER lumen by an endogenous signal peptide. In ER, GPC is post translationally cleaved at a conserved WAASA site, and two glycoproteins G1 and G2 are generated which are later glycosylated and translocated to the Golgi. Glycoproteins facilitate the attachment of virions with the integrin receptors located on the host cell surface.

RdRp

Hnatavirus RdRp is a huge protein with a molecular weight of 250-280 KD. Because of its large molecular weight RdRp is difficult to express in bacteria, and thus remains the most uncharacterized protein in hantaviruses. RdRp mediates both transcripn and replication of viral genome. During transcription RdRp synthesizes viral mRNA from negative sense vRNA template. During replication RdRp replicates vRNA genome via a cRNA intermediate. Thus it is likely that hantavirus RdRp has multiple activities, including endonuclease, replicase, transcriptase and RNA helix unwinding activities. However, recent studies have shown that viral RdRp requires N protein for function. For example, short capped primers generated from host cell mRNAs by the process of cap snatching are used by RdRp to initiate the transcription. Hnatavirus N protein has been found to be involved in the generation of such capped primers (21).

Epidemiology

The geographic distribution of hantavirus hosts mirrors the hantavirus epidemiology and geographic distribution (Table1). Hantavirus infection to humans is considered a spill over infection that causes two types of serious illnesses, HFRS and HPS. The primary root of infection for both these illnesses is the inhalation of live virus through lungs. In general humans get hantavirus infection by direct contact with infected rodents or their aerosolized excreta; although there are reports documenting the spread of Andes virus (ANDV) from human to human (8, 23). HFRS is caused by old world hantaviruses and most of its cases are found in Eastern Asia (China, Korea and Eastern part of Russia) and Europe, including the European part of Russia). Annually more than 100,000 HFRS cases are reported in China alone (15), and over 900 cases are reported in Korea and Eastern Russia (19). In Europe majority of HFRS cases are registered in Russia, Finland and Sweden (Fig 4, table 1). Majority of HFRS patients are males with age of 20 to 50 years. HFRS mortality rate depends upon the type of virus and in general varies from 0.1 to 10%. HRFS patients mostly belong to rural areas where the hantaviral rodent hosts are thickly inhabited. The only hantavirus that causes diseases in urban areas is the Seoul virus (SEOV) because its host is a domestic rat (Rattus norvegicus and Rattus rattus). HPS has a mortality of 40-50% and is caused by new world hantaviruses, including SNV, ANDV, Monongahela virus, New York virus, Black Creek Canal virus, Bayou virus, Oran virus and numerous other newly identified strains. Although HPS is found throughout united states, the majority of cases are registered in western region and are caused by SNV. In fact SNV is the predominantly found viral species that causes HPS in patients. HPS out breaks in North America are associated with increased population of host deer mouse (peromyscus maniculatus). HPS has also been reported in other countries in south and central America, including Argentina, Brazil, Chili, Bolivia, Paraguay, Uruguay and Panama.

Figure 4.

Figure 4

World wide distributation of hantavirus species

Pathogenesis

It is not yet exactly clear how hantaviruses spread the human body after their inhalation through lungs. However, immature Dendritic cells (DCs) likely play a significant role in their dissemination. Immature DCs express β3 integrins, the receptors that hantaviruses use for attachment and entry. In the airways and alveoli of lungs, the DCs located in the vicinity of epithelial cells serve as primary targets for pathogen pick up. Hantaviruses infect both immature and mature DCs, which likely serve as vehicles for the transport of virions through the lymphatic vessels to the regional lymph nodes, where they get opportunity to infect other immune cells, such as, macrophages and monocytes. After further replication the free or cell bound virions can infect the endothelial cells, the ultimate targets for viruses causing hemorologic fever (20).

Increased vascular permeability and decreased platelet count are the hallmarks of hantavirus associated disease, and the mechanics of such pathogenesis is poorly understood. Unlike other hemorologic fever viruses such as Ebola virus, the hantaviruses do not increase the permeability of endothelial cell monolayers in vitro, nor do they cause any cytopathic effects in the host endothelial cells. This points towards the role of host immune system in hantavirus pathogenesis.

Invading viruses are detected early during infection by non immune cells as well as DCs located at host pathogen interface (2). Pathogen recognition receptors (PRRs) for virus detection in host cells include TLRs and RIG-I like helicases (RIG-I and MDA5). RIG-I detects viruses from multiple families, including Orthomyxoviridae, paramyxoviridae, Rhabdoviridae and Flaviviridae. However, the PRRs that sense hantaviruses in host cells remains to be identified. Type I INF and other pro-inflammatory responses triggered by PRR signaling induce host resistance for viral infection and activation of innate immune cells, such as, natural killer (NK) cells or NKT cells for host defense. These early responses are aimed to reduce the viral dissemination during lag phase before adaptive immune response is ready to strike. It is well established that treatment of cells with type I INF induces the expression of several hundred INF stimulating genes (ISGs), including the well characterized MxA protein that has antiviral activity against the viruses of the Bunyaviridae family (9, 17). Interestingly, most pathogenic hantaviruses have evolved the strategies to sabotage the INF induced host defense mechanisms. For example, pathogenic hantaviruses (HTNV, NYV) delay the type-I INF response, including MxA expression in endothelial cells. This virus programmed delay in the establishment of antiviral state in host cells generates a window of opportunity for rapid replication and spread of pathogenic hantaviruses through endothelial cell layer.

Inflammatory cytokines/chemokines produced by antiviral innate immune response represent a double edged sword. On one hand, they play a role in virus elimination, but on other hand, if not properly regulated they can facilitate a virus associated disease. For example, TNFα can purge viruses from infected cells with out causing cell lysis. However, at the same time TNFα can modulate the endothelial barrier functions by promoting vascular leakage by increasing leukocyte adhesion and transendothelial migration (3). TNFα and other cytokines are suspected to play role in Ebola virus (EBOV) mediated septic shock during EBOV hemerologic fever. Although hantavirus infection to DCs in vitro produces substantial amount of TNFα, which is consistent with increased plasma levels of this cytokine in hantavirus infected patients during acute phase of HFRS. However, there is no evidence of such a harmful innate immune response, producing a “cytokine storm” similar to 1918 strain of influenza virus, that could contribute to the hantavirus immunopathogenesis.

In general, DCs are the prime targets for viruses to evade immune system. For example, human cytomegalovirus and herpes simplex viruses (DNA viruses) impair the function of DCs and induce their apoptosis by different mechanisms (18). Filoviruses, such as, EBOV and Marburgvirus infect immature human DCs and inhibit their transition to antigen presenting cells and also impair their ability to produce T-cell stimulatory cytokines (25). On the other hand, hantaviruses infect immature DCs and activate their maturation and transition into antigen presenting cells with out causing any cytopathic effects. Mature DCs after hantavirus infection had increased T-cell stimulatory capacity, explaining the elicitation of vigorous T-cell response with long lasting memory in patients during acute phase of hantavirus infection (35). Although epitopes are defined for all three hantavirus proteins, N protein is the major viral target antigen recognized by T cells, likely due to its higher comparative abundance in infected cells. In recent years the concept that CTL response directed against hantavirus infected endothelial cells mediates increased capillary permeability is gaining more and more support from multiple indirect evidences. For example, T cell attracting chemokines CCL5 and CXCL10 are secreted by human lung microvascular endothelial cells after infection by either HTNV or SNV in vitro (31). Hantavirus specific CTLs efficiently lyse human endothelial cells leading to the increased vascular permeability (12). In addition, PUUV infected patients show a peak CTL response at the onset of disease with increased serum levels of perforin, granzyme B, and the epithelial cell apoptosis marker caspase-cleaved cytokeratin-18 (16, 32). The number of CD8+ T cells in PUUV infected patients strongly increase during the acute phase of HFRS, and leave a strong long-lived CTL memory against the PUUV N protein (35). It is likely that a strong unusual primary response accounts for such a long-lived hantavirus specific CTL memory. These observations suggest that the strength of antiviral CTL response and the number of available CTL targets in endothelial cell layer could determine the severity of damage to the vascular bed in hantavirus infected patients. An interesting question that how do the reservoir host rodents escape the vascular damage during persistent hantavirus infection remains to be answered. However, recent studies have suggested that regulatory T cells may play an important role in limiting immunopathology in the natural reservoir host, but this response may interfere with viral clearance. It is hypothesized that in humans the immune mechanisms that downregulate the hantavirus specific CTL response and facilitate the viral clearance by non-cytolytic means are missing, leading to a CTL storm for rapid elimination of virus at the expense of costly damage to the endothelial barrier that causes fatal capillary leakage. In contrast, rodent hosts have elevated regulatory T cell response that controls the CTL activity, leading to viral persistence without immunopathology (Fig 5). The quality of T cell response trigerred against hantaviruses is determined by DCs, which are programmed through PRRs during early stages of infection. Thus, further investigation is required to identify the PRRS that detect hantaviruses in rodent hosts and humans. It is probable that PRRS for hantaviruses in humans and rodents are different and program DCs differently that leads to a fatal disease in humans and viral persistence in rodent hosts. Similar to cellular immunity, hantaviruses induce a stable and long lasting humoral immune response involving antibodies of all Ig subclasses (IgA, IgM, IgG). Antibodies against N protein appear soon after the onset of the disease and antibodies against G1 and G2 appear later during the progress of the disease. High titers of antibodies have been found in individuals that have experienced HCPS years ago. Humans infected with PUUV show the presence of neutralizing antibodies in blood decades post infection, suggesting that previously infected individuals are protected life long from the infection. Tables 2 and 3 summarize the immune response in humans and rodents by hantavirus infection.

Figure 5.

Figure 5

Working hypothesis of differential regulation of hantavirus-specific immune responses in rodent reservoir hosts and humans. During their encounter with viruses DCs integrate different signals received through several PRRs (PRR 1, PRR 2, PRR3, etc.) which determine the quality of the ensuing T-cell response. (A) In their rodent reservoir host, hantavirus-associated PRR signaling could program DCs to stimulate Treg cells that can suppress virus-specific CTLs, leading to viral persistence and at the same time preventing virus-induced immunopathology. (B) In humans, who are not adapted to hantaviruses, PRR signaling in DCs results in a dominant antiviral CTL response. As a consequence, hantavirus-infected endothelial cells (EC) are immediately eliminated leading to immunopathology.

Note: This picture was obtained from Ref (29)

Table 2.

Summary of immune response in humans during hantavirus infection

Categorical
Response
Immune Marker Effect on
infection
Virus species a In vitro /
In vivo
Tissue or Cell type b
Phase of infection c

Innate RIG-I Elevated SNV In vitro HUVEC, ≤ 24 h p.i
Reduced NY-I In vitro HUVEC, ≤ 24 h p.i
TLR3 Elevated SNV In vitro HUVEC, ≤ 24 h p.i
IFN-β Elevated PUUV, PHV,
ANDV
In vitro HSVEC, HMVEX, , ≤ 24 h
p.i
Reduced TULV, PUUV NSs In vitro COS-7 and MRC5 cells, ≤
24 h p.i
INF-α Elevated PUUV, HTNV In vitro Mφ, DCs, 4 days p.i
No change HTNV In vitro Blood, acute
IRF-3, IRF-7 Elevated SNV, HTNV, PHV In vitro HMVEC-L, , ≤ 24 h p.
MxA Elevated HTNV, NY-IV,
PHV, PUUV,
ANDV, SNV,
In vitro Mφ, HUVEC, HMVEC-L,
6h-4 days p.i
MCH I & II Elevated HTNV In vitro DCs, 4 days p.i
CD 11b Elevated PUUV In vitro Blood, acute
CD-40, CD80,
CD86
Elevated HTNV In vitro Dcs, 4 days p.i
Nk cells Elevated PUUV In vitro Bal, acute

Proinflammatory /
Adhesion
IL-1β Elevated SNV, HTNS In vitro Blood, lungs, acute
IL-6 Elevated SNV, PUUV In vitro Blood, lungs, acute
NF-α Elevated PUUV, SNV,
HTNV
In vitro Blood, lungs, acute, kidney
CCL5 Elevated SNV, HTNV In vitro HMVEC-L, HUVEC, 3-4
days p.i
CXCL8 Elevated PUUV In vitro Blood, acute
Elevated PUUV In vitro Men, blood, acute
Elevated TULV, PHV,
HTNV
In vitro HUVEC, Mφ, 4-2 days p.i
CXCL10 Elevated SNV, HTNV, PHV In vitro HUVEC-L, HUVEC, 3-4
days p.i
Elevated PUUV In vitro Men, blood, acute
IL-2 Elevated SNV, HTNV,
PUUV
In vitro Blood, lungs, acute
Nitric Oxide Elevated PUUV In vitro Blood, acute
GM-CSF Elevated PUUV In vitro Women, blood, acute
ICAM, VCAM Elevated PUUV In vitro Kidney acute
Elevated HTNV, PHV In vitro HUVEC, 3-4 days p.i
E- selectin Elevated PUUV In vitro Blood, acute

CD8+ and
CD4+ T cells
INF-γ Elevated HTNV, SNV In vitro Blood, CD4+, CD8+,
lunges, acute
CD8+ Elevated DOBV, PUUV,
HTNV
In vitro Blood, BAL, acute
Virus specific INF-
γ+CD8+
Elevated PUUV, SNV In vitro PBMC, acute
Perforin,
GranzymeB
Elevated PUUV In vitro Blood acute
CD4+CD25+
“activated”
Elevated DOB, PUUV In vitro PBMC, acute
IL-4 Elevated SNV In vitro Lungs, acute

Regulatory Suppressor
T cells d
Reduced HTNV In vitro Blood, acute
IL-10 Elevated PUUV In vitro Blood, acute
TGF- β Elevated PUUV In vitro Kidney, acute

Humoral IgM, IgA, IgG, IgE Elevated All hantaviruses In vitro Blood

Note: Contents of this table are from reference (6)

a

SNV, Sin Nombre virus; NY-IV, New York -1; PUUV, Puumala viris; PHV, Prospect Hill virus; ANDV, Andes virus; TULV, Tula virus; HTNV, Hantaan virus; DOBV, Dobrava virus

b

HUVEC, human umbilical vascular endothelial cells, HSVEC, human saphenous vein endothelial cells; HMVEC-L, human lung microvascular endothelial cells; COS-7, African green monkey kidney fibroblast transformed with Siman virus 40; MRC5, human fetal lung fibroblasts; Mφ, Macrophages; DCs, dendritic cells; Bal, bronchoalveolar lavage, PMBC, human peripheral blood mononuclear cells.

c

Acute infection is during symptomatic disease in patients.

d

Suppressor T cells likely represent cells currently referred to as regulatory T cells

Table 3.

Summary of immune response in rodents during hantavirus infection

Categorical
Response
Immune
Marker
Effect on
infection
Virus
species a
Host, Tissue or Cell type b Phase of infection c

Innate TLR7 Redeuced SEOV Male Norway rats, lungs Acute, Persistent
Elevated SEOV Female Norway rats, lungs Acute, Persistent
RIG-I Elevated SEOV Female Norway rats, lungs Acute, Persistent
Elevated SEOV Norway rats, Thalamus Acute
TLR3 Elevated SEOV Male Norway rats, lungs Acute, Persistent
INF-β Reduced SEOV Male Norway rats, lungs Acute, Persistent
Elevated SEOV Female Norway rats, lungs Acute
MX2 Reduced SEOV Male Norway rats, lungs Acute, Persistent
Elevated SEOV Female Norway rats, lungs Acute, Persistent
Elevated HTN,
SEOV
Mice d, fibroblast transfected
with MX2
3-4 days p.i
JAK2 Elevated SEOV Female Norway rats, lungs Acute
MHC II Elevated PUUV, Bank Voles Genetic susceptibility

Proinflammatory /
Adhesion
IL-1β Reduced SEOV, Male Norway rats, lungs Persistent
IL-6 Reduced SEOV Male & female Norway rats,
lungs
Acute, Persistent
Elevated SEOV Male rats spleen Acute
TNF-α Reduced HTNV Newborn mice d, CD8+, Spleen Acute
Reduced SEOV Male Norway rats, lungs Acute persistent
Elevated SEOV Female Norway rats, lungs Acute
CXCL1 / Reduced SEOV Male Norway rats, lungs Acute persistent
CXCL10 Elevated SEOV Male Norway rats, Spleen Acute
CCL2 /
CCL5
Elevated SEOV Male Norway rats, Spleen Acute
NOS2 Reduced SEOV Male Norway rats, lungs Acute persistent
Elevated SEOV Male Norway rats, spleen Acute
Elevated HTNV Mouse Mφ d, In vitro 6 h p.i
VCAM /
VGF
Elevated SEOV Male Norway rats, spleen Acute

CD8+ and
CD4+ T cells
CD8+ Reduced HTNV, Newborn mice d, spleen Persistent
Elevated HTNV, SCID mice d, CD8+ transferred Persistent
Elevated SEOV Female Norway rats, lungs Persistent
INF-γ Elevated SEOV Female Norway rats, lungs Acute
Elevated SEOV Male Norway rats, lungs Acute
Elevated SEOV Female & male Norway rats Acute
Elevated SNV Deer mice, CD4+ T cells Acute
Elevated HTNV Newborn mice d CD4+ T cells Acute
Reduced HTNV Newborn mice d CD4+ T cells Persistent
INF-γR Elevated SEOV Female Norway rats, lungs Acute, persistent
Reduced SEOV Male Norway rats, lungs Persistent
T cells Elevated SEOV Nude rat Persistent
Elevated HTNV Nude mice d Persistent
IL-4 Reduced SEOV Male Norway rats, lungs Acute, persistent
Elevated SNV Deer mice, CD4+ T cells Acute
Elevated SEOV Female & male Norway rats Acute

Regulatory Regulatory
T cells
Elevated SEOV Male Norway rats, lungs Persistent
FoxP3 Elevated SEOV Male Norway rats, lungs Persistent
TGF- β Elevated SEOV Male Norway rats, lungs Persistent
SNV Deer mice, CD4+ T cells Persistent
IL-10 Reduced SEOV Male Norway rats,lungs, spleen Persistent
Elevated SNV Deer mice, CD4+ T cells Persistent

Humoral IgG Elevated SNV Deer mice Persistent
Elevated SEOV Norway rats Persistent
Elevated HTNV Field mice Persistent
Elevated PUUV Banl Voles Persistent
Elevated BCCV Cotton rats Persistent

Note: Contents of this table are from reference (6)

a

SEOV, Seoul virus; SNV, Sin Nombre virus; HTNV, Hantaan virus; PUUV, Puumala virus; BCCV, Black Creek Canal virus.

b

Mφ, Macrophages.

c

Acute infection is < 30 days p.i and persistent infection is ≥ 30 days p.i.

d

Mus musculus, non-natural reservoir host for hantaviruses

Clinical presentation

As described by the Centers for Disease Control and Prevention (CDC), Patients with HCPS typically present a short febrile prodrome of 3-5 days (31). In addition to fever and myalgias, early symptoms include headache, chills, dizziness, non-productive cough, nausea, vomiting, and other gastrointestinal symptoms. Malaise, diarrhea, and lightheadedness are reported by approximately half of all patients, with less frequent reports of arthralgias, back pain, and abdominal pain. Patients may report shortness of breath, (respiratory rate usually 26 - 30 times per minute). Typical findings on initial presentation include fever, tachypnea and tachycardia, with usually a normal physical exam.

The analysis of clinical, laboratory, and autopsy data of seventeen patients with confirmed hantavirus infection during 1993 hantavirus outbreak reveled the mean duration of symptoms before hospitalization is 5.4 days. The most common symptoms at the time of hospitalization were fever, myalgia, headache, cough, and nausea or vomiting (Tables 4 and 5), with myalgia being the most frequently reported initial symptom. The most common physical findings were tachypnea and tachycardia (TablesTables 6 and 7). Fifty percent of the patients had a respiratory rate of 28 or more breaths per minute, and 50 percent had a heart rate of 120 or more beats per minute. No patient had conjunctival hemorrhage, petechial rash, clinical signs of internal hemorrhage (including a guaiac-positive stool specimen), or peripheral or periorbital edema. Notable hematologic findings included an elevated white-cell count with increased neutrophils, myeloid precursors, and atypical lymphocytes

Table 4.

Symptoms in 17 Patients with Hantavirus Infection

Symptom No of patients (%)
Fever 17(100)
Myalgia 17(100)
Headache 12(71)
Cough 12(71)
Nausea or vomiting 12(71)
Chills 11(65)
Malaise 10(59)
Diarrhea 10(59)
Shortness of breath 9(53)
Dizziness or lightheadedness 7(41)
Arthralgia 5(29)
Back pain 5(29)
Abdominal pain 4(24)
Chest pain 3(18)
Sweats 3(18)
Dysuria or frequent urination 3(18)
Rhinorrhea or nasal Congestion 2(12)
Sore throat 2(12)

Note: Contents of this table are from reference (5)

Table 5.

Symptoms in 10 pediatric patients (age ≤16 years) with SNV Infection

Symptom No of patients (%)
Fever 10(100)
Myalgia 8(80)
Headache 10(100)
Cough 9(90)
Nausea or vomiting 9(90)
Chills 3(30)
Diarrhea 4(40)
Shortness of breath 8(80)
Dizziness or lightheadedness 3(30)
Back pain 5(50)
Abdominal pain 5(50)
Chest pain 3(30)
Sore throat 4(40)

Note: Contents of this table are from reference (26)

Table 6.

Clinical findings at the time of admission in 17 Patients with hantavirus Infection

Sign Percentage of patients Median (Range)
Respiratory rate ≥20/min 100 28 (20-70)
Heart rate ≥100 bpm 94 120 (90-150)
Temperature ≥38.1 °C 75 38.8 (35.4-40.4)
Systolic or rales on lung examination 31
Abdominal tenderness 24
Cool, Clammy, or mottled skin 18
Injection or suffusion of conjunctiva 18

Note: Contents of this table are from reference (5)

Table 7.

Clinical findings at the time of admission in 9 pediatric patients with SNV infection.

Sign Percentage of patients
Tachypnea* 67
Fever (temperature ≥38.0 °C) 56
Crackles or rales on lung exam 44
Abdominal tenderness 44
Hypotension@ 33
Tachycardia (heart rate >120 bpm) 22
Cool, Clammy, or mottled skin 11

Resoiratory rate >25 breats / min (10-13 years old). Respiratory rate > 20 breaths / min (≥14 years old). Includes 2 patients mechanically ventilated before admission.

@

Systolic blood pressure <95 mm Hg (10-13 years old). Systolic blood pressure <100 mm Hg ((≥14 years old)

Note: Contents of this table are from reference (26)

The partial-thromboplastin time was 40 seconds or longer in 67 percent of the patients at the time of admission (TablesTables 8 and 9). Although minimal abnormalities of renal function were common, the serum creatinine levels did not rise above 2.5 mg per deciliter (220 μmol per liter) in any patient (Tables 8 and 9). The mean specific gravity of urine at the time of admission was 1.024 ±0.010. Forty percent of patients had proteinuria on admission. Urine dipstick tests for blood were positive in fifty seven percent of tested patients at the time of admission.

Table 8.

Results of laboratory studies during hospitalization in patients with hantavirus Infection

Test* Admission value Maximal [Minimal]
Value
White cells-×103 /mm3 10.4(3.1-65.3) 26.0 (5.6-65.3)
Band forms - % 22 (8-62) 27 (4-67)
Hematocrit -%
   Men 51.3 (49.9-60.0) 56.3 (49.9-67.7)
   Women 46.4 (35.0-55.8) 48.5 (36.5-60.3)
Platelets – ×103 /mm3 84 (26-320) [64] (12-148)
Prothrombin time – sec 13.0 (11.2-21.1) 14 (12.6-21.1)
Partial-thromboplastin time – sec 42.5 (30.0-150.0) 54.4 (31.0-150.0)
Bicarbonate – mmol / liter 18 (12-25) [14] (8-20)
Lactate dehydrogenase IU / liter 362 (209-1525) 568 (324-1525)
Aspartate aminotransferase IU / liter 112 (28-432) 148 (62-432)
Alanine aminotransferase IU / liter 55 (25-148) 63 (27-149)
Albumin – g / dl 3.0 (1.5-4.6) [2.5] (1.5-3.5)
Blood urea nitrogen – g / dl 11 (3-23) 17 (8-32)
Creatinine – mg / dl 1.1 (0.6-2.5) 1.4 (0.6-2.5)
Lactate – mmol / liter 4.4 (2.2-11.0) -
Creatine kinase – 1I / liter 46 (19-1026) -
*

Maximal normal values: Lactate dehydrogenase, 180 to 232 IU per liter; aspartate aminotransferase, 35 to 40 IU perliter; alanine aminotransferase, 35 to 60 IU per liter, Lactate, 2.2 mmol per lire; and creatine kinase, 180 to 269 IU per liter. To convert values fro creatinine to micro moles per liter, multiply by 88.4

@

Two of ten patients tested had elevated creatne kinase concentrations: 1026 IU per liter with an MB fraction of 16 (2 percent) in 1 patient, and 814 IU per liter with an MB fraction of 87 (11 percent) in another patient, who was undergoing cardiopulmonary resuscitation when the sample was obtained.

Note: Contents of this table are from reference (5)

Table 9.

Results of laboratory studies at the time of admission in pediatric patients with Sin Nombre hantavirus infection.

Test Admission value
(Median [Range])
White cells × 103 /mm3 9.0 (3.4-59.2)
Mematocrit %
   Patients 10-12 y (n = 5) 42.0 (34.9-45.2)
   Males 13-16 y (n = 2) 54.3 (47.6-61)
   Females 13-16 y (n = 4) 41.9 (40-48.4)
Platelets × 103 /mm3 67 (43-98)
Creatinine (mg / dl) 0.7 (0.4-3.9)
Prothrombin time (sec) 13.1 (11.0-29.8)
Partial thromboplastin time (sec) 38(27-212)
Carbon dioxide (mmol / liter) 20 (15-27)
Blood urea nitrogen (mg / dl) 10 (8-26)
Aspartate aminotransferase (IU / L) 98 (39-129)
Albumin (g / dl) 2.8 (1.2-3.5)
Alanine aminotransferase (IU / L) 55 (21-80)
Lactate dehydrogenase (IU / L) 1243 (382-1724)
Lactate (mmol / L) 2.5 (1.5-18.4)

Note: Contents of this table are from reference (26)

The initial chest radiograph showed interstitial or interstitial and alveolar infiltrates (Fig 6) in sixty five percent, fluffy alveolar infiltrates in twelve percent, and no abnormalities in twenty four percent of the patients. Subsequently, ninety four percent had rapidly evolving, bilateral, diffuse infiltrates, and six percent had interstitial infiltrates confined to the lower lobes. Pleural effusions were noted during the course of the illness in four patients. Eleven of twelve patients (ninety two percent) who underwent chest radiography and arterial measurement of oxygen saturation at the time of admission had either pulmonary infiltrates or arterial oxygen saturation under 90 mm Hg. Similar observations were made in other hantavirus infected cases (1, 24)

Figure 6.

Figure 6

Radiographs showing the evolution of hantavirus pulmonary syndrome in a 30-year-old woman. (A) Chest radiograph before onset of illness. (B) Admission radiograph. (C) Radiograph after intubation. (D) Radiograph just before death.

Note: This picture was obtained from Ref (11)

Since the emergence of HCPS in United States in 1993, only 5.5% of the cases reported to CDC were children less than sixteen years of age. Due to minimum causalities in younger children and adolescents, it is hypothesized that hantavirus infection in children is less likely to develop into serious illness as compared to adults. The analysis of clinical data of ten SNV pediatric cases is presented in Tables 4-9.

Diagnostics

Hnatavirus infection is diagnosed on the basis of a positive serological test and the confirmation of viral antigen in the tissue of infected patient or the presence of viral RNA sequences in patient’s blood or tissue, along with a compatible history of the disease.

Serologic assays

During 1993 hantavirus outbreak, cross-reactive antibodies to the previously known hantaviruses, such as, Hantaan, Seoul, Puumala, and Prospect Hill virus were found in the acute- and convalescent-phase sera of some HPS patients. Since then, tests based on specific viral antigens from SNV have been developed and are widely used for the routine diagnosis of HPS. Enzyme-linked immunosorbent assay (ELISA) is the popular test for the detection of IgM antibodies in patient’s blood that are raised against Hantaviruses during infection.

An IgG test in conjunction with the IgM-capture test is also used for the diagnosis of Hantavirus disease. Acute and convalescent-phase sera should reflect a fourfold rise in IgG antibody titer or the presenc of IgM in acute-phase sera for a positive hantavirus infection. It may be noted that acute-phase serum used as an initial diagnostic specimen may not yet have IgG. IgG is a long lasting antibody, retained for many years after infection. Thus, SNV IgG ELISA has been used in serologic investigations of the epidemiology of the disease and appears to be appropriate for this purpose. Rapid immunoblot strip assay (RIBA) is an investigational prototype assay for the identification of serum antibodies to recombinant proteins and peptides specific for SNV and other hantaviruses. Also, neutralizing plaque assays have recently been performed for the serological confirmation of SNV infections. However, these specific assays are not commercially available. Isolation of hantaviruses from human sources is difficult and no isolates of SNV-like viruses have been recovered from humans. Thus, isolation of hantavirus is not considered for diagnostic purposes.

Immunohistochemistry (IHC)

IHC testing of formalin-fixed tissues with specific monoclonal and polyclonal antibodies can be used to detect hantavirus antigens and has proven to be a sensitive method for laboratory confirmation of hantaviral infections. IHC has an important role in the diagnosis of HPS in patients from whom serum samples and frozen tissues are unavailable for diagnostic testing and in the retrospective assessment of disease prevalence in a defined geographic region.

Polymerase Chain Reaction (PCR)

Reverse transcriptase polymerase chain reaction (RT-PCR) is a very sensitive assay and can be used for the detection of hantaviral RNA in infected samples, such as, lung tissues and blood clots from infected patients. However, RT-PCR is very prone to cross-contamination and should be considered an experimental technique with a limited use for diagnostic purposes of hantavirus infections.

Differential diagnosis

A variety of Infectious etiologies, such as, pneumonia, sepsis with ARDS, and acute bacterial endocarditis can often be confused with HPS. Other conditions commonly found in the southwest United States have presentations similar to HPS, such as, septicemic plague, tularemia, histoplasmosis, and coccidioidomycosis. In addition, noninfectious conditions, including myocardial infarction with pulmonary edema and Goodpasture’s syndrome should also be considered.

Treatment

Apart from supportive care there is no treatment for hantavirus infection at present. Patients receive broad spectrum antibiotics while awaiting the results of laboratory diagnostic tests for the confirmation of hantavirus associated disease. Initial supportive care includes the use of antipyretics and analgesics. Patients are immediately transferred to intensive care unit (ICU) if preliminary symptoms indicate the higher probability of HPS. ICU management should include careful assessment, monitoring and adjustment of volume status and cardiac function, including inotropic and vasopressor support if needed. Fluids should be administrated carefully due to higher chances of capillary leakage. Supplemental oxygen is necessary for hypoxic patients. Due to high risks of respiratory failure, ICU management should keep equipment and materials for intubation and mechanical ventilation readily available. Patients with severe HPS quickly progress to respiratory failure, and in absence of ECMO (extracorporeal membrane oxygenation) almost all patients die within 24-48 hours of the onset of this severe phase.

Antiviral therapy including the use of ribavirin, a guanosine analogue, has not been shown to be effective for the treatment of HPS. However, efficacy trials in HFRS patients in China have shown significant beneficial effects of ribavirin if started early in the disease course. Although ribovarin perturbs SNV replication in vitro, neither an open-label trial conducted during the 1993 outbreak nor an attempted placebo-controlled trial demonstrated clinical benefit for HPS. However, it has been suggested that ribavirin efficacy may depend on phase of infection and the severity of the disease at the time of administration. Ribavirin is not recommended for treatment of HPS and is not available for this use.

Examination of neutralizing antibody titers in patients at the time of admission has revealed low antibody titers for the patients with severe disease and higher antibody titers for patients with mild disease. These observations provide clues towards the use of human neutralizing antibodies during the acute phase of HPS that might prove effective for the treatment and / or prophylaxis of hantaviral infections.

There is no FDA-approved vaccine for hantavirus infection in USA. A killed-virus vaccine against hantavirus infection has been developed in Korea and China. However, such approaches are not persuaded in USA due to many reasons. Efforts are underway to develop a DNA vaccine, based on the gene gum approach for the transport of an expression plasmid containing M segment gene to the individual, which upon expression will generate neutralizing antibodies against glycoproteins G1 and G2.

Immunity and reinfection

High titers of neutralizing antibodies have been found in individuals who have experienced HPS years ago (34, 36). Neutralizing antibodies against PUUV have been detected in individuals decades after infection (30). These observations suggest that previously infected individuals are protected life-long from reinfection. There are no known re-infections with the homologous hantavirus. Closely related hantaviruses, such as Seoul and Hantaan viruses, seem to cross-protect against re-infection in experimental animals, and one might expect cross-protection among the hantaviruses derived from sigmodontine rodents.

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 citable 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.

References

  • 1.Center for Disease Control and Prevention Case definitions for infectious conditions under public health surveillance. MMWR Morb Mortal Wkly Rep. 1997;46:1–55. [PubMed] [Google Scholar]
  • 2.Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998;392:245–52. doi: 10.1038/32588. [DOI] [PubMed] [Google Scholar]
  • 3.Bradley JR. TNF-mediated inflammatory disease. J Pathol. 2008;214:149–60. doi: 10.1002/path.2287. [DOI] [PubMed] [Google Scholar]
  • 4.Carey DE, Reuben R, Panicker KN, Shope RE, Myers RM. Thottapalayam virus: a presumptive arbovirus isolated from a shrew in India. Indian J Med Res. 1971;59:1758–60. [PubMed] [Google Scholar]
  • 5.Duchin JS, Koster FT, Peters CJ, Simpson GL, Tempest B, Zaki SR, Ksiazek TG, Rollin PE, Nichol S, Umland ET, et al. Hantavirus pulmonary syndrome: a clinical description of 17 patients with a newly recognized disease. The Hantavirus Study Group. N Engl J Med. 1994;330:949–55. doi: 10.1056/NEJM199404073301401. [DOI] [PubMed] [Google Scholar]
  • 6.Easterbrook JD, Klein SL. Immunological mechanisms mediating hantavirus persistence in rodent reservoirs. PLoS Pathog. 2008;4:e1000172. doi: 10.1371/journal.ppat.1000172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Elliott RM, Schmaljohn CS, Collett MS. Bunyaviridae genome structure and gene expression. Curr Top Microbiol Immunol. 1991;169:91–141. doi: 10.1007/978-3-642-76018-1_4. [DOI] [PubMed] [Google Scholar]
  • 8.Enria D, Padula P, Segura EL, Pini N, Edelstein A, Posse CR, Weissenbacher MC. Hantavirus pulmonary syndrome in Argentina. Possibility of person to person transmission. Medicina (B Aires) 1996;56:709–11. [PubMed] [Google Scholar]
  • 9.Frese M, Kochs G, Feldmann H, Hertkorn C, Haller O. Inhibition of bunyaviruses, phleboviruses, and hantaviruses by human MxA protein. J Virol. 1996;70:915–23. doi: 10.1128/jvi.70.2.915-923.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Gavrilovskaya IN, Gorbunova EE, Mackow NA, Mackow ER. Hantaviruses direct endothelial cell permeability by sensitizing cells to the vascular permeability factor VEGF, while angiopoietin 1 and sphingosine 1-phosphate inhibit hantavirus-directed permeability. J Virol. 2008;82:5797–806. doi: 10.1128/JVI.02397-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Graziano KL, Tempest B. Hantavirus pulmonary syndrome: a zebra worth knowing. Am Fam Physician. 2002;66:1015–20. [PubMed] [Google Scholar]
  • 12.Hayasaka D, Maeda K, Ennis FA, Terajima M. Increased permeability of human endothelial cell line EA.hy926 induced by hantavirus-specific cytotoxic T lymphocytes. Virus Res. 2007;123:120–7. doi: 10.1016/j.virusres.2006.08.006. [DOI] [PubMed] [Google Scholar]
  • 13.Johnson KM. Hantaviruses: history and overview. Curr Top Microbiol Immunol. 2001;256:1–14. doi: 10.1007/978-3-642-56753-7_1. [DOI] [PubMed] [Google Scholar]
  • 14.Khaiboullina SF, Morzunov SP, St Jeor SC. Hantaviruses: molecular biology, evolution and pathogenesis. Curr Mol Med. 2005;5:773–90. doi: 10.2174/156652405774962317. [DOI] [PubMed] [Google Scholar]
  • 15.Khan A, Khan AS. Hantaviruses: a tale of two hemispheres. Panminerva Med. 2003;45:43–51. [PubMed] [Google Scholar]
  • 16.Klingstrom J, Hardestam J, Stoltz M, Zuber B, Lundkvist A, Linder S, Ahlm C. Loss of cell membrane integrity in puumala hantavirus-infected patients correlates with levels of epithelial cell apoptosis and perforin. J Virol. 2006;80:8279–82. doi: 10.1128/JVI.00742-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Kochs G, Janzen C, Hohenberg H, Haller O. Antivirally active MxA protein sequesters La Crosse virus nucleocapsid protein into perinuclear complexes. Proc Natl Acad Sci U S A. 2002;99:3153–8. doi: 10.1073/pnas.052430399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Larsson M, Beignon AS, Bhardwaj N. DC-virus interplay: a double edged sword. Semin Immunol. 2004;16:147–61. doi: 10.1016/j.smim.2004.02.002. [DOI] [PubMed] [Google Scholar]
  • 19.Lee HW. Hemorrhagic fever with renal syndrome in Korea. Rev Infect Dis. 1989;11(Suppl 4):S864–76. [PubMed] [Google Scholar]
  • 20.Marty AM, Jahrling PB, Geisbert TW. Viral hemorrhagic fevers. Clin Lab Med. 2006;26:345–86. viii. doi: 10.1016/j.cll.2006.05.001. [DOI] [PubMed] [Google Scholar]
  • 21.Mir MA, Duran WA, Hjelle BL, Ye C, Panganiban AT. Storage of cellular 5′ mRNA caps in P bodies for viral cap-snatching. Proc Natl Acad Sci U S A. 2008;105:19294–9. doi: 10.1073/pnas.0807211105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Mir MA, Panganiban AT. A protein that replaces the entire cellular eIF4F complex. EMBO J. 2008;27:3129–39. doi: 10.1038/emboj.2008.228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Padula PJ, Edelstein A, Miguel SD, Lopez NM, Rossi CM, Rabinovich RD. Epidemic outbreak of Hantavirus pulmonary syndrome in Argentina. Molecular evidence of person to person transmission of Andes virus. Medicina (B Aires) 1998;58(Suppl 1):27–36. [PubMed] [Google Scholar]
  • 24.Bruno D. L. H. Philip, MD, Brown Joel, MD, Tanner William, MD, Lau Alan., MD The protean Manifestations of Hemorrhagic Fever with Renal Syndrome. Annals of Internal Medicine. 1990;113:385–391. doi: 10.7326/0003-4819-113-5-385. [DOI] [PubMed] [Google Scholar]
  • 25.Pollara G, Kwan A, Newton PJ, Handley ME, Chain BM, Katz DR. Dendritic cells in viral pathogenesis: protective or defective? Int J Exp Pathol. 2005;86:187–204. doi: 10.1111/j.0959-9673.2005.00440.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Ramos MM, Overturf GD, Crowley MR, Rosenberg RB, Hjelle B. Infection with Sin Nombre hantavirus: clinical presentation and outcome in children and adolescents. Pediatrics. 2001;108:E27. doi: 10.1542/peds.108.2.e27. [DOI] [PubMed] [Google Scholar]
  • 27.Schmaljohn C, Hjelle B. Hantaviruses: a global disease problem. Emerg Infect Dis. 1997;3:95–104. doi: 10.3201/eid0302.970202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Schmaljohn CS. Bunyaviridae: The viruses and their replication. In: Fields BN, Knipe DM, Howley PN, editors. Fields Virology. Lippencott-Raven; Philadelphia: 1996. pp. 1581–602. [Google Scholar]
  • 29.Schonrich G, Rang A, Lutteke N, Raftery MJ, Charbonnel N, Ulrich RG. Hantavirus-induced immunity in rodent reservoirs and humans. Immunol Rev. 2008;225:163–89. doi: 10.1111/j.1600-065X.2008.00694.x. [DOI] [PubMed] [Google Scholar]
  • 30.Settergren B, Ahlm C, Juto P, Niklasson B. Specific Puumala IgG virus half a century after haemorrhagic fever with renal syndrome. Lancet. 1991;338:66. doi: 10.1016/0140-6736(91)90069-2. [DOI] [PubMed] [Google Scholar]
  • 31.Sundstrom JB, McMullan LK, Spiropoulou CF, Hooper WC, Ansari AA, Peters CJ, Rollin PE. Hantavirus infection induces the expression of RANTES and IP-10 without causing increased permeability in human lung microvascular endothelial cells. J Virol. 2001;75:6070–85. doi: 10.1128/JVI.75.13.6070-6085.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Tuuminen T, Kekalainen E, Makela S, Ala-Houhala I, Ennis FA, Hedman K, Mustonen J, Vaheri A, Arstila TP. Human CD8+ T cell memory generation in Puumala hantavirus infection occurs after the acute phase and is associated with boosting of EBV-specific CD8+ memory T cells. J Immunol. 2007;179:1988–95. doi: 10.4049/jimmunol.179.3.1988. [DOI] [PubMed] [Google Scholar]
  • 33.Vaheri A, Vapalahti O, Plyusnin A. How to diagnose hantavirus infections and detect them in rodents and insectivores. Rev Med Virol. 2008;18:277–88. doi: 10.1002/rmv.581. [DOI] [PubMed] [Google Scholar]
  • 34.Valdivieso F, Vial P, Ferres M, Ye C, Goade D, Cuiza A, Hjelle B. Neutralizing antibodies in survivors of Sin Nombre and Andes hantavirus infection. Emerg Infect Dis. 2006;12:166–8. doi: 10.3201/eid1201.050930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Van Epps HL, Terajima M, Mustonen J, Arstila TP, Corey EA, Vaheri A, Ennis FA. Long-lived memory T lymphocyte responses after hantavirus infection. J Exp Med. 2002;196:579–88. doi: 10.1084/jem.20011255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Ye C, Prescott J, Nofchissey R, Goade D, Hjelle B. Neutralizing antibodies and Sin Nombre virus RNA after recovery from hantavirus cardiopulmonary syndrome. Emerg Infect Dis. 2004;10:478–82. doi: 10.3201/eid1003.020821. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES