Ebola and Marburg haemorrhagic fever viruses: Major scientific advances, but a relatively minor...

15
Ebola and Marburg haemorrhagic fever viruses: major scientific advances, but a relatively minor public health threat for Africa E. M. Leroy 1,2 , J-P Gonzalez 1 and S. Baize 3 1) Centre International de Recherches Me ´dicales de Franceville, Franceville, Gabon, 2) MIVEGEC (IRD 224, CNRS 5290, Universite ´s Montpellier 1), Institut de Recherche pour le De ´veloppement, Montpellier, France and 3) Unite ´ de Biologie des Infections Virales Emergentes, Institut Pasteur, IFR128-Biosciences Gerland-Lyon Sud, Lyon, France Abstract Ebola and Marburg viruses are the only members of the Filoviridae family (order Mononegavirales), a group of viruses characterized by a linear, non-segmented, single-strand negative RNA genome. They are among the most virulent pathogens for humans and great apes, causing acute haemorrhagic fever and death within a matter of days. Since their discovery 50 years ago, filoviruses have caused only a few outbreaks, with 2317 clinical cases and 1671 confirmed deaths, which is negligible compared with the devastation caused by malnutrition and other infectious diseases prevalent in Africa (malaria, cholera, AIDS, dengue, tuberculosis ). Yet considerable human and financial resourses have been devoted to research on these viruses during the past two decades, partly because of their potential use as bioweapons. As a result, our understanding of the ecology, host interactions, and control of these viruses has improved consid- erably. Keywords: Ebola, immunity, Marburg, outbreak, reservoir, vaccine Clin Microbiol Infect Corresponding author: E. M. Leroy, Centre International de Re- cherches Me ´dicales de Franceville, Franceville, Gabon E-mail: [email protected] Introduction Ebola (EBOV) and Marburg (MARV) are the only members of the Filoviridae family and are among the most virulent pathogens for humans and great apes. The most virulent spe- cies of these viruses induce acute haemorrhagic fever and death within a few days in up to 90% of symptomatic individ- uals [1,2]. The explosive disease course, high case fatality rate, and lack of specific treatments or vaccines make EBOV and MARV a major public health issue for Africa, and a cate- gory A biothreat [3,4]. In the field, clinical specimens must be handled according to WHO guidelines on viral haemor- rhagic fever agents in Africa [5], and analyzed in biosafety level 4 laboratories. Research on filoviruses has been closely tied to outbreak activity. Some advances in the biology and pathogenesis of MARV and EBOV were made just after the discovery of these agents, in 1967 and 1976, respectively. The two viruses were then largely neglected until EBOV re-emerged in 1995 in Kik- wit, Democratic Republic of Congo (DRC), where it caused a large outbreak that received considerable media attention worldwide. The extraordinarily high lethality, multifocal haem- orrhaging, and specificity to African ecosystems enhanced the fascination of Ebola for the international community, which saw this disease as a potential threat in the post-Cold War context [6]. This partly explains why research on the biology, epidemiology, ecology and pathophysiology of these viruses has advanced markedly over the past 15 years, leading recently to the development of potential antiviral drugs and candidate vaccines. Yet the overall disease burden of filovirus- es over the past 45 years is minimal compared with major infectious diseases such as AIDS, malaria, cholera, dengue and tuberculosis [7,8]. This paper reviews research advances in Ebola and Marburg ecology, virus–host interactions and dis- ease control during the past 15 years. C L M 3 5 3 5 B Dispatch: 19.4.11 Journal: CLM CE: Nathiya Journal Name Manuscript No. Author Received: No. of pages: 13 PE: Gayathri ª2011 The Authors Clinical Microbiology and Infection ª2011 European Society of Clinical Microbiology and Infectious Diseases REVIEW 10.1111/j.1469-0691.2011.03535.x 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50

Transcript of Ebola and Marburg haemorrhagic fever viruses: Major scientific advances, but a relatively minor...

Ebola and Marburg haemorrhagic fever viruses: major scientific

advances, but a relatively minor public health threat for Africa

E. M. Leroy1,2, J-P Gonzalez1 and S. Baize3

1) Centre International de Recherches Medicales de Franceville, Franceville, Gabon, 2) MIVEGEC (IRD 224, CNRS 5290, Universites Montpellier 1), Institut

de Recherche pour le Developpement, Montpellier, France and 3) Unite de Biologie des Infections Virales Emergentes, Institut Pasteur, IFR128-Biosciences

Gerland-Lyon Sud, Lyon, France

Abstract

Ebola and Marburg viruses are the only members of the Filoviridae family (order Mononegavirales), a group of viruses characterized by

a linear, non-segmented, single-strand negative RNA genome. They are among the most virulent pathogens for humans and great

apes, causing acute haemorrhagic fever and death within a matter of days. Since their discovery 50 years ago, filoviruses have caused

only a few outbreaks, with 2317 clinical cases and 1671 confirmed deaths, which is negligible compared with the devastation caused by

malnutrition and other infectious diseases prevalent in Africa (malaria, cholera, AIDS, dengue, tuberculosis …). Yet considerable human

and financial resourses have been devoted to research on these viruses during the past two decades, partly because of their potential

use as bioweapons. As a result, our understanding of the ecology, host interactions, and control of these viruses has improved consid-

erably.

Keywords: Ebola, immunity, Marburg, outbreak, reservoir, vaccine

Clin Microbiol Infect

Corresponding author: E. M. Leroy, Centre International de Re-

cherches Medicales de Franceville, Franceville, Gabon

E-mail: [email protected]

Introduction

Ebola (EBOV) and Marburg (MARV) are the only members

of the Filoviridae family and are among the most virulent

pathogens for humans and great apes. The most virulent spe-

cies of these viruses induce acute haemorrhagic fever and

death within a few days in up to 90% of symptomatic individ-

uals [1,2]. The explosive disease course, high case fatality

rate, and lack of specific treatments or vaccines make EBOV

and MARV a major public health issue for Africa, and a cate-

gory A biothreat [3,4]. In the field, clinical specimens must

be handled according to WHO guidelines on viral haemor-

rhagic fever agents in Africa [5], and analyzed in biosafety

level 4 laboratories.

Research on filoviruses has been closely tied to outbreak

activity. Some advances in the biology and pathogenesis of

MARV and EBOV were made just after the discovery of these

agents, in 1967 and 1976, respectively. The two viruses were

then largely neglected until EBOV re-emerged in 1995 in Kik-

wit, Democratic Republic of Congo (DRC), where it caused a

large outbreak that received considerable media attention

worldwide. The extraordinarily high lethality, multifocal haem-

orrhaging, and specificity to African ecosystems enhanced the

fascination of Ebola for the international community, which

saw this disease as a potential threat in the post-Cold War

context [6]. This partly explains why research on the biology,

epidemiology, ecology and pathophysiology of these viruses

has advanced markedly over the past 15 years, leading

recently to the development of potential antiviral drugs and

candidate vaccines. Yet the overall disease burden of filovirus-

es over the past 45 years is minimal compared with major

infectious diseases such as AIDS, malaria, cholera, dengue and

tuberculosis [7,8]. This paper reviews research advances in

Ebola and Marburg ecology, virus–host interactions and dis-

ease control during the past 15 years.

CL

M3

53

5B

Dispa

tch:

19.4.11

Jour

nal:

CLM

CE:N

athiya

JournalName

ManuscriptNo.

Autho

rRec

eive

d:No.

ofpa

ges:

13PE

:Gay

athr

i

ª2011 The Authors

Clinical Microbiology and Infection ª2011 European Society of Clinical Microbiology and Infectious Diseases

REVIEW 10.1111/j.1469-0691.2011.03535.x

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

21

22

23

24

25

26

27

28

29

30

31

32

33

34

35

36

37

38

39

40

41

42

43

44

45

46

47

48

49

50

Filoviruses as Prototypes of EmergingPathogens

An emerging pathogen is defined as either a truly novel or a

hitherto unknown agent, or a pathogen whose incidence has

increased considerably in susceptible populations. EBOV and

MARV fulfil one criterion of this definition, in that they were

identified relatively recently. The first reported cases of

human filovirus infection occurred in 1967 in Marburg, Ger-

many, and in Belgrade, Serbia (former Yugoslavia), when lab-

oratory technicians were infected while manipulating African

green monkeys (Cercopithecus aethiops) imported from

Uganda [9–12]. An unknown infectious agent was recovered

from the victims’ blood and organs and was named ‘Marburg

virus’ after the outbreak location. EBOV first emerged in the

form of two near-simultaneous outbreaks in 1976 in Sudan,

near the border with DRC, and in DRC, near the border

with Sudan [13,14]. An unknown causative agent was isolated

from patients in the DRC outbreak and was named for the

river Ebola, which flows past Yambuku, the outbreak epicen-

tre (Fig. 1).

The morphology of MARV and EBOV is unique among

viruses (Fig. 2a,b). Using electron microscopy, they often

appear as long filamentous particles about 14 000 nm long

and 80 nm wide, whereas other particles assume the shape

of the number 6 or a hairpin [15–17]. MARV and EBOV are

the only members of the Filoviridae family. This family belongs

to the order Mononegavirales, which also includes Rhabdoviri-

dae, Paramyxoviridae and Bornaviridae, a group of viruses

characterized by a linear, non-segmented, single negative-

stranded RNA genome [18–23].

These viruses cause an explosive disease with multiple

haemorrhaging occurring within a few days and a high fatality

rate (up to 90%), which was rarely observed in viral diseases

[24–29]. Generally, after an incubation period ranging from 2

to 21 days (mean 4–9 days), non-specific symptoms such as

fever, headache, nausea and muscle pain occur abruptly.

There are rapidly followed by gastrointestinal problems

(stomach pain, vomiting, diarrhoea), respiratory disorders

(throat and chest pain, cough) and neurological manifesta-

tions (prostration, confusion, delirium), indicating systemic

viral dissemination and multi-organ failure. Haemorrhagic

manifestations vary in severity and location, and often include

skin rash, conjunctival injection, nosebleeds, melaena, hae-

matemesis and bleeding at venepuncture sites. In fatal cases

the symptoms and haemorrhaging rapidly become severe,

and death generally ensues in a context of tachypnoea,

coma and shock. The virus disappears rapidly from the

bloodstream after the resolution of systemic symptoms in

survivors, and prolonged convalescence is frequent, with epi-

sodic non-specific symptoms such as asthenia, myalgia and

fever. More specific signs, associated with transient persis-

tence of the virus in immunologically protected sites such as

the eyes and testicles have been reported during the early

stages of recovery. The virus has been isolated for up to

2 months in semen [30–32]. Biological parameters are not

specific for filovirus infection and most data come from

experimentally infected animals [33,34]. They include mild

and early leucopenia, associated with lymphopenia, thrombo-

cytopenia (< 100 000 platelets/mm3), hyperproteinaemia, and

marked transaminase elevation. The prothrombin time is

always prolonged and fibrin split products are often

detected, indicating disseminated intravascular coagulation

[35,36].

(a)

(b)

Republic

of Congo

2001–02

2003

2005

Democratic

Republic of Congo

1976

1995

2007

2008

Sudan

1976

1979

2004

Uganda

2000

Ivory Coast 1995

Uganda

2007

Gabon

1994

1996

1996–97

2001–2002

Kenya

1980

1987

South Africa

1975

Democratic

Republic of Congo

1998–2000

Angola

2005

Uganda

2007

2008

CIEBOV

BEBOV

SEBOV

ZEBOV

Marburg

Ebola

FIG. 1. Filovirus outbreaks in Africa. Reported outbreaks or isolated

cases of haemorrhagic fever caused by Marburg virus (MARV) (a)

and Ebola virus (EBOV) (b) are indicated with the corresponding

year and colour-coded according to virus species.

COLOR

2 Clinical Microbiology and Infection CMI

ª2011 The Authors

Clinical Microbiology and Infection ª2011 European Society of Clinical Microbiology and Infectious Diseases, CMI

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

21

22

23

24

25

26

27

28

29

30

31

32

33

34

35

36

37

38

39

40

41

42

43

44

45

46

47

48

49

50

Filoviruses: a Questionable Public HealthBurden for Africa

Since their discovery 45 years ago, filoviruses have been

responsible for only a few outbreaks, resulting in 2317 clini-

cal cases and 1671 confirmed deaths (reviewed in Fig. 1 and

Table 1, [37]). Hence, the disease burden of filovirus infec-

tion in Africa is extremely small compared with other infec-

tious diseases and malnutrition.

As said previously, the first MARV outbreak occurred in

Europe and was associated with 31 cases, including 25 pri-

mary cases (i.e. infected directly by animals) with seven

deaths [9–11]. Since then, only sporadic cases have been

reported, in South Africa in 1975 (three cases, one death)

and in Kenya in 1980 (two cases, one death) and 1987 (one

fatal case) [31,38,39]. MARV subsequently caused two large

outbreaks, in 1998–2000 in DRC and in 2004–2005 in

Angola. The longest MARV outbreak hit the Durba area

(eastern DRC) between 1998 and 2000, affecting 154 people

and killing 83% of victims [40]. Most patients worked in

underground gold mines. The Durba outbreak was charac-

terized by the circulation of multiple viral strains, pointing to

multiple independent introductions from the unknown

(a) (b)

(c)

VP40

VP24

NPVP30RNA

VP35LMembrane

GP1,2

trimer

(d)

FIG. 2. 10Ultrastructure and structure of filovirus virons. Particles of various forms are visualized by negative staining by uracyl acetate (a, b). Part

of a rod-shaped particle showing the tubular nucleocapsid and the surface spikes (c). Schematic representation of a filovirion indicating protein

composition, the tubular nucleocapsid and the surface spikes (d). (From Kuhn JH 9. Filoviruses: a compendium of 40 years of epidemiological, clini-

cal, and laboratory studies. In: Calisher CH editor. Springer, Wien, NewYork; 2008).

TABLE 1. List of filovirus outbreaks with the corresponding

location, number of clinical cases and fatality rate

Virus Date Location CasesFatality(%)

Marburg virus 1967 Germany – Yugoslavia 31 231975 South Africa 3 331980 Kenya 2 501987 Kenya 1 100

1998–2000 DRC 154 832005 Angola 252 902007 Uganda 4 252007 Uganda 1 02008 Uganda 1 100

Ebola virusZaire

1976 DRC 318 881977 DRC 1 1001994 Gabon 49 651995 DRC 315 881996 Gabon 37 57

1996–1997 Gabon 60 752001–2002 Gabon and

Republic of Congo123 79

2003 begin Republic of Congo 143 902003 end Republic of Congo 35 83

2005 Republic of Congo 11 752007 DRC 264 712008 DRC 32 47

Ebola virusSudan

1976 Sudan 286 531979 Sudan 34 652000 Uganda 425 532004 Sudan 17 42

Ebola virusCote d’Ivoire

1994 Ivory Coast 1 0

Ebola virusBundibugyo

2007 Uganda 102 42

DRC, Democratic Republic of Congo.

LOW

RESOLUTIO

NCOLOR

FIG

CMI Leroy et al. Ebola and Marburg haemorrhagic fever viruses1

3

ª2011 The Authors

Clinical Microbiology and Infection ª2011 European Society of Clinical Microbiology and Infectious Diseases, CMI

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

21

22

23

24

25

26

27

28

29

30

31

32

33

34

35

36

37

38

39

40

41

42

43

44

45

46

47

48

49

50

natural reservoir [29,40,41]. The largest recorded MARV

outbreak occurred in Uige province of northern Angola in

2004–2005 [42]. It was the first recorded outbreak in West

Africa, causing 252 cases and 227 deaths (fatality rate 90%).

Complete genomic characterization showed that the culprit

strain was closely related to previous East African isolates

[42]. Finally, small MARV outbreaks occurred in western

Uganda in 2007 and 2008 among workers in lead and gold

mines of the Kitaka Cave near Ibanda village [43,44]. Two of

the six victims died.

Ebola virus first emerged in the form of two near-simulta-

neous outbreaks in 1976 caused by two different species,

namely Sudan Ebola virus (SEBOV) in Sudan and Zaire Ebola

virus (ZEBOV) in DRC [13,14]. In Sudan, the outbreak was

centred in the Nzara and Maridi and was responsible for 284

cases. The mortality rate was 53%, characteristic of SEBOV

infection. The epicentre of the DRC outbreak was in Yamb-

uku, about 800 km from Maridi. This outbreak resulted in

318 cases with a mortality rate of 89%, characteristic of

ZEBOV infection. Later there was an unconfirmed lethal case

involving a 9-year-old girl living in Tandala, DRC, [45], fol-

lowed by another SEBOV outbreak in 1979, again in Nzara

and Maridi, with 34 cases and 22 deaths [46]. After a 15-year

period in which no further cases were recorded, EBOV

re-emerged in 1994 for a 3-year period. This new phase was

marked by the identification of a new species, Cote d’Ivoire

Ebola virus (CIEBOV), and by four ZEBOV outbreaks. The

only case caused by CIEBOV occurred in 1994, when an eth-

nologist sickened a few days after autopsying a chimpanzee

found dead in the Tai national park in Ivory Coast [47–49].

A large outbreak then occurred in 1995, in and around the

town of Kikwit, south of DRC, with 315 cases and a mortal-

ity rate of 81% [50]. Despite the deployment of more

sophisticated scientific and medical resources than those

available in 1976, this outbreak was as large as the 1976 out-

break, probably because it affected a town with several hun-

dred thousand inhabitants. Three other outbreaks, all as the

result of ZEBOV, struck northeast Gabon between 1994 and

1997 [51–54]. In total, these outbreaks involved 140 cases

and caused 75 deaths. The first occurred in gold-digger

camps located in the heart of the forest. The second out-

break hit the village of Mayibout, located south of Mekouka,

among children who had carried and butchered a chimpan-

zee carcass found in the forest. The third outbreak occurred

in 1996–1997, with 60 cases and 45 deaths over a 6-month

period. Fifteen cases and 11 deaths were recorded in Libre-

ville, the capital of Gabon, and a South African nurse was

infected by a Gabonese physician who had travelled to

Johannesburg. The period 2000–2008 was marked by

repeated ZEBOV outbreaks, SEBOV resurgence, and the dis-

covery of a new species of Ebola virus, Bundibugyo Ebola

virus (BEBOV). Between 2001 and 2005, Gabon and Republic

of Congo (RC) were hit by five ZEBOV outbreaks [55–58].

The first outbreak occurred within the cross-border area

between northeast Gabon and northwest RC, and resulted

in 143 cases and 128 deaths. RC was again affected twice in

2003 (respectively 143 and 35 cases, and 128 and 29 deaths),

and then in 2005 at Etoumbi, where only 11 cases (nine fatal-

ities) were reported. Two outbreaks of Sudan Ebola virus

also occurred during this period. One hit Uganda in 2000,

comprised three foci and was the largest of all recorded

EBOV outbreaks, causing 173 deaths among its 425 victims

[59,60]. A second SEBOV outbreak occurred in Sudan in

2004 in the town of Yambio, located near Nzara and Maridi.

There were 17 cases and seven deaths [61–63]. The latest

species to be discovered, Bundibugyo Ebola virus (BEBOV),

was discovered in 2007 in Uganda, where it was responsible

for a large outbreak [64] with 116 cases and 30 deaths (fatal-

ity rate 26%).

These four EBOV species are all pathogenic for humans

and arose in sub-Saharan Africa. An additional species, Res-

ton Ebola virus (REBOV), was isolated in 1989 from Asian

cynomolgus monkeys (Macaca fascicularis) imported from the

Philippines and housed in a quarantine facility in Reston, Vir-

ginia, USA. The monkeys developed a haemorrhagic disease

associated with high lethality, and subsequent outbreaks

among cynomolgus monkeys were reported in animal facili-

ties in Texas, USA, Italy and the Philippines [39,65–68].

Although no humans fell sick during these episodes, several

monkey handlers in the United States and in the Philippines

were shown to have seroconverted [69,70]. Recently, REB-

OV was also isolated from domestic Philippino swine with a

severe respiratory syndrome and co-infected by porcine

reproductive and respiratory syndrome virus [71].

Filoviral Genes and Proteins

The MARV and EBOV genomes are about 19 000 nucleo-

tides long and are transcribed into eight major subgenomic

mRNAs, which encode seven structural proteins: 3¢ leader,

nucleoprotein (NP), virion protein 35 (VP35), VP40, glyco-

protein (GP), VP30, VP24, and RNA-dependent RNA poly-

merase (L)-5¢ trailer; and one non-structural protein (sGP),

shown in Fig. 2. The central core of the virion is composed

of the ribonucleoprotein complex, which consists of the

genomic RNA molecule encapsulated by NP linked to the

inner matrix proteins VP30 and VP35 and the RNA-depen-

dent RNA polymerase (Fig. 2d). This complex is involved in

transcription and replication [72–74]. The matrix proteins

4 Clinical Microbiology and Infection CMI

ª2011 The Authors

Clinical Microbiology and Infection ª2011 European Society of Clinical Microbiology and Infectious Diseases, CMI

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

21

22

23

24

25

26

27

28

29

30

31

32

33

34

35

36

37

38

39

40

41

42

43

44

45

46

47

48

49

50

VP40 and VP24 are linked to the ribonucleoprotein complex

at the inner surface of the lipid bilayer of the viral envelope,

which is derived from the host cell. VP40 and VP24 are

involved in viral nucleocapsid formation, viral budding or

assembly, and host range determination [72,75–82]. The viral

envelope contains only the glycoproteins, organized as tri-

meric spikes (Fig. 2c,d) consisting of two fragments (extracel-

lular protein, GP1, and membrane-anchored protein, GP2)

which result from furin-like enzyme cleavage of precursor

polyproteins and remain linked by a disulphide bond [74,83–

85]. GP binds preferentially to endothelial and monocytic

cells via GP1, mediating target cell entry, inducing endothelial

cell disruption and cytotoxicity in blood vessels in vitro, and

causing immunosuppression in vitro via a retrovirus-like pep-

tide [74,86–94]. EBOV differs from MARV and from other

members of the Mononegavirales in that infected cells

secrete large amounts of a non-structural glycoprotein

(sGP), the primary product of the GP gene, which is

expressed from non-edited mRNA species [93,95,96]. The

role of sGP is poorly understood. Finally, the matrix proteins

VP35 and VP24 seem to play a key role in the pathogenicity

of EBOV and MARV, by inhibiting antiviral responses and

particularly type 1 interferon (IFN) synthesis [97–102].

Filoviruses Genomics

The Filoviridae family comprises two genera, Marburgvirus and

Ebolavirus, and belongs to the order Mononegavirales, a

group of viruses characterized by a genome consisting of a

linear, non-segmented, single negative strand of RNA [18–

23]. Molecular evolutionary analyses of the glycoprotein

genes suggests that EBOV and MARV diverged several thou-

sand years ago [103]. The genus Marburgvirus consists of a

single species, Marburgvirus, comprising five viruses differing

from one another by up to 21% at the nucleotide level, while

there are five known species of Ebolavirus that have different

geographic locations, case fatality rates, and nucleotide

sequence divergences of about 32–41% [16].

Our knowledge of filovirus distribution and diversity may

be limited because EBOV and MARV have only been identi-

fied and characterized in sick or dead humans and animals

[40,42,43,53–56,64,68,71,104–107]. Numerous studies have

attempted to determine the regions of EBOV and MARV cir-

culation. Remotely sensed data and niche ecologic modelling

suggest that filoviruses may circulate within a much wider

region than their outbreak areas [108–111]. Using a sensitive

and specific ELISA method, an overall ZEBOV-specific IgG se-

roprevalence of 15.3% was found in Gabon, the highest ever

reported [112]. The seroprevalence was significantly higher

in forested areas (19.4%) than in grassland (12.4%), savannah

(10.5%) and areas rich in lakes (2.7%). These results are con-

sistent with previous reports of seroprevalence rates ranging

from 1.8% to 21.3% throughout central Africa using poorly

specific immunofluorescence antibody testing 2methods [113–

117]. More convincingly, two recent small serosurveys, based

on the same ELISA as the one we used, have shown high

antibody prevalence in some forested regions of Central

Africa. Indeed, a 9.3% (15/161) prevalence of ZEBOV-specific

IgG was found in unaffected villages surrounding Kikwit a few

weeks after the 1995 outbreak [118], and a 13.2% (25/190)

seroprevalence was found in the Aka Pygmy population in

the Central African Republic, where no ZEBOV outbreaks

have been reported [119]. Such high seroprevalence rates

indicate a high level of ZEBOV circulation in the tropical rain

forest environment and suggest that human rural populations

are highly exposed to the virus, with mild or asymptomatic

infection by outbreak strains [120] or less pathogenic strains,

or simple exposure to inert viral particles.

Filoviruses in Wildlife

Filovirus haemorrhagic fevers are typical zoonotic diseases

transmitted accidentally by direct contact with live or dead

animals. The role of wildlife species in the human epidemiol-

ogy of ZEBOV is only partly understood (Fig. 3). In addition

to its high pathogenicity for humans, ZEBOV has caused

massive outbreaks among gorillas and chimpanzees, killing

thousands of animals during the last decade in parts of Ga-

bon and RC [56,121–125]. Another study conducted in the

Tai forest of Ivory Coast showed that the disappearance of

11 members (26%) of a group of 43 chimpanzees during

November 1994 may have been the result of CIEBOV [126].

Such massive outbreaks in great apes could have major impli-

cations for animal conservation. The identification of multiple

strains during the 2001 Gabon/RC outbreak and the recent

identification of two phylogenetically divergent lineages sug-

gest independent introductions into great ape and human

populations following multiple viral spill-overs from a reser-

voir host [56,107,127]. In this ‘multi-emergence’ hypothesis,

Ebola outbreaks would occur episodically in certain ecologi-

cal conditions caused by habitat disturbances or climatic phe-

nomena [108,109]. Although the idea of multi-emergence

makes no reference to a particular timescale, this theory also

implicitly assumes that ZEBOV was present in Equatorial

Africa long before the first documented outbreak in 1976, as

supported by various serosurveys [128–130].

Although many field and laboratory studies have been

conducted since the first recorded outbreak in an attempt

CMI Leroy et al. Ebola and Marburg haemorrhagic fever viruses1

5

ª2011 The Authors

Clinical Microbiology and Infection ª2011 European Society of Clinical Microbiology and Infectious Diseases, CMI

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

21

22

23

24

25

26

27

28

29

30

31

32

33

34

35

36

37

38

39

40

41

42

43

44

45

46

47

48

49

50

to identify the reservoir species of MARV and EBOV, conclu-

sive evidence that bats are natural hosts for filoviruses was

obtained only recently [14,45,131–138]. Antibodies and

nucleotide sequences specific for ZEBOV [127,139] were

detected in the liver and spleen of three fruit bat species in

Gabon and RC (Hypsignathus monstrosus, Epomops franquetti

and Myonycteris torquata), and antibodies and nucleotide

sequences specific for MARV [140,141] were found in a fruit

bat species in Gabon (Rousettus aegyptiacus) and in two insec-

tivorous bat species in DRC (Rhinolophus eloquens and Mini-

opterus inflatus). More recently, MARV was isolated from

cave-dwelling Rousettus aegyptiacus bats in Uganda [43].

Finally, a recent study showed that the 2007 Luebo outbreak

in DRC was linked to massive fruit bat migration, strongly

suggesting that humans could be infected directly by bats

(Fig. 3) [142].

Pathogenesis of Ebola Virus Infection andImmune Responses

Antigen-presenting cells such as dendritic cells and macro-

phages are major EBOV targets [143,144]. Indeed, by their

presence in mucosal tissues and skin, these cells are infected

early and serve as the substrate for the first replicative viral

cycles. Because of their extensive distribution among the dif-

ferent organs and tissues, and their mobility, dendritic cells

and macrophages are probably responsible for viral dissemi-

nation and systemic infection [145]. Intense viral replication

then occurs in secondary lymphoid organs and liver, and the

virus subsequently spreads to hepatocytes, endothelial cells,

fibroblasts and some epithelial cells [146,147]. The pathogen-

esis of filovirus infection is reviewed in Fig. 4.

However, despite widespread viral replication, the

observed damage to endothelia, liver and other organs is not

sufficiently severe to cause terminal shock and death. Endo-

thelial infection is infrequent, occurring in the terminal stages

with no clear evidence of cytopathic effects [36]. Similarly,

although multifocal hepatic necrosis may be linked to viral

replication in liver, the observed hepatocellular lesions can-

not directly lead to death [146,147]. A pathogenic role of

the host response is therefore suspected.

Extensive infection of monocytes and macrophages leads to

aberrant release of inflammatory mediators and chemokines

such as interleukin-1b (IL-1b), tumour necrosis factor-a (TNF-

a), IL-6, IL-15, IL-16, IL-1 receptor antagonist, soluble TNF

receptor, IL-10, NO), IL-8, growth regulated oncogene-a,

CCL3, CCL4, CXCL10, monocyte chemotactic protein-1, and

eotaxin [144,148–151]. This ‘cytokine storm’, particularly

prominent in the terminal stages of the disease in humans and

non-human primates, has dramatic consequences. It is proba-

ble that TNF-a, NO), and other vasoactive compounds play a

crucial role in the associated vascular leakage, by increasing

endothelial permeability, reducing vascular tone, and altering

endothelial cell functions [144,152–154]. In addition, some of

these mediators may induce the expression of adhesion mole-

cules at the surface of endothelial cells, so allowing neutroph-

ils and monocytes to invade sites of infection where they may

cause bystander tissue damage [155]. Infected macrophages

are involved in the coagulopathy and in the induction of dis-

seminated intravascular coagulation by their abundant expres-

sion of tissue factor [35,156,157]. Other consequences of the

massive antigen-presenting cell infection are lymphoid deple-

tion in the spleen, lymph nodes and thymus, and extensive

apoptosis of T lymphocytes and natural killer cells, as

observed in blood and tissues of human and non-human pri-

mate fatalities [145,150,154,158–161]. T-cell death cannot be

the result of a direct viral cytopathic effect, as lymphocytes

are not infected by EBOV, but would rather result from inter-

actions with infected antigen-presenting cells and soluble medi-

Reservoirs of ZEBOV & MARV:

fruit bats species

Intermediary susceptible species:

virus amplification in great apes and duikers

Outbreak

Suspected

Direct

contact

FIG. 3. 11Model of the natural cycle of filovirus. The diagram shows animal-to-human transmissions leading to outbreak appearance. Several Zaire

Ebola virus (ZEBOV) outbreaks appeared when hunters handled the infected carcasses of chimpanzee, gorilla and duiker. Several ZEBOV and

Marburg virus (MARV) outbreaks were suspected to be associated with exposure to fruit bats.

LOW

RESOLUTIO

NCOLOR

FIG

6 Clinical Microbiology and Infection CMI

ª2011 The Authors

Clinical Microbiology and Infection ª2011 European Society of Clinical Microbiology and Infectious Diseases, CMI

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

21

22

23

24

25

26

27

28

29

30

31

32

33

34

35

36

37

38

39

40

41

42

43

44

45

46

47

48

49

50

ators [36,150,158]. A viral protein with superantigen activity

might also be involved [162]. Although T-cell depletion proba-

bly induces profound immunosuppression, the consequences

for disease progression are unclear [163,164]. Fatal Ebola

haemorrhagic fever is also associated with defective humoral

responses, with absent specific IgG and barely detectable IgM

[158,165]. This impairment of adaptive immunity may result

from extensive infection of dendritic cells. Indeed, EBOV-

infected dendritic cells produce only a limited panel of cyto-

kines without full activation and maturation, which may impair

their ability to trigger adaptive immunity [143,145,166,167].

Some viral factors are also able to counteract innate

immunity and thereby impede the control of viral replication.

Indeed, VP35 blocks IFN-a/b synthesis by preventing the acti-

vation of interferon regulatory factors 3 and 7

[97,99,101,102,168]. VP35 also interferes with the activation

of dsRNA-dependent protein kinase [169], and, like VP30

and VP40 [170], is a suppressor of RNA silencing [171].

VP24 impairs the nuclear accumulation of tyrosine-phosphor-

ylated signal transducer and activator of transcription 1 and

thereby inhibits IFN-a/b and IFN-c signalling [172,173]. Inhibi-

tion of type I IFN responses seems to be crucial for EBOV

virulence, as viruses that have lost this property by VP35

mutation are attenuated in vitro and in vivo [174,175].

In striking contrast with fatal outcome, effective control of

EBOV infection is associated with balanced immune

responses. A transient and well-regulated inflammatory

response is observed early in the disease not only in patients

that survive, but also in asymptomatic subjects [120,149].

This response may help to control the infection and to

induce specific immunity. An early and robust humoral

response is strongly correlated with survival in symptomatic

patients [158,165]. In asymptomatic EBOV-infected subjects,

antibodies are detected about 3 weeks after infection and in

moderate amounts [120]. In survivors and asymptomatic sub-

jects, high antibody titres persist for years [176]. Although

FIG. 4. A model of pathogenesis of filovirus infection, based on findings with Zaire Ebola virus. Dendritic cells and macrophages are early targets

of filoviruses. The virus spreads from the initial site of infection to secondary lymphoid organs and liver where intense replication takes place.

Then, other cells are infected such as hepatocytes, endothelial cells, fibroblasts and some epithelial cells. The inhibition of type I interferon (IFN)

production by viral proteins leads to relentless viral replication in most organs. The extensive infection of antigen-presenting cells (APC) leads to

altered inflammatory response and uncontrolled release of mediators. This ‘cytokine storm’ contributes to the pathogenesis by attracting inflam-

matory cells towards infected tissues, inducing coagulopathy and increasing endothelial permeability and vascular leakage. In addition, infected

APC and/or soluble factors are responsible for the defective adaptive immunity and the massive apoptosis of T lymphocytes. Multifocal necrosis

of hepatocytes, liver failure, destruction of secondary lymphoid organs and other tissue damage may result from direct cytopathic effects but also

from the host response. Together, these events lead to multi-organ failure, impairment of the vascular system, terminal shock and death.

COLOR

CMI Leroy et al. Ebola and Marburg haemorrhagic fever viruses1

7

ª2011 The Authors

Clinical Microbiology and Infection ª2011 European Society of Clinical Microbiology and Infectious Diseases, CMI

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

21

22

23

24

25

26

27

28

29

30

31

32

33

34

35

36

37

38

39

40

41

42

43

44

45

46

47

48

49

50

antibodies probably participate in the control of infection,

their precise role is unclear. Importantly, control of EBOV

infection is also characterized by a lack of T-cell apoptosis

and by the circulation of activated T cells (probably including

cytotoxic T lymphocytes), coinciding with a fall in viraemia

and the disappearance of symptoms [158]. A similar

response also occurs in asymptomatic individuals [177], sug-

gesting that T-cell responses are crucial for controlling the

infection. The factors underlying the radically different out-

comes of EBOV infection are unclear, but may include the

route of infection, the size of the inoculum, the cell types

that are initially infected, previously acquired immunity [112],

heterologous immunity, and MHC status [178]. However, it

is unlikely that outcomes were related to different virus

strains, as no difference was found in the coding sequences

of viruses isolated from survivors and fatalities and different

outcomes can be observed in patients infected with the same

viral source [51,105].

Treatment and Prevention

Although several advances in the treatment and prevention

of EBOV infection have recently been described, there is no

licensed vaccine, and the only way to limit outbreaks is to

isolate patients. The development of therapeutic measures is

hampered not only by the need to manipulate EBOV in bio-

safety level 4 facilities, but also by the limited number of

people affected since the discovery of filoviruses. However,

the increasing frequency of EBOV outbreaks in humans and

great apes [56,122–124] and the recent emergence of new

EBOV species [47,64], give evidence for the intense circula-

tion of EBOV in the endemic area.

The main challenge is to develop a vaccine conferring

cross-protection against heterologous EBOV species, includ-

ing emerging strains, ideally after a single dose. Several vac-

cine candidates have shown good efficacy in non-human

primates over the last 10 years. The use of live viral vectors

or virus-like particles to produce EBOV GP is a promising

approach, providing sterilizing immunity after a single dose

[179–184]. Recent studies have demonstrated the feasibility

of a multivalent vaccine able not only to protect against the

filovirus species used to formulate the candidate

[180,185,186] but also to cross-protect against an emerging

species [187]. However, several concerns must be dealt with

before human use, such as the safety of the viral vector, pos-

sible pre-existing immunity to the vector, and formulation of

a single-dose cross-protective vaccine. Results obtained with

candidate vaccines confirm the major role of T-cell

responses, and of CD8+ T cells in particular, in the control

of filovirus infections, as well as the presence of cross-react-

ing immunogenic epitopes on the viral GP.

A vesicular stomatitis virus-based candidate also has thera-

peutic potential, conferring substantial post-exposure protec-

tion when administered very rapidly to non-human primates

[188,189]. Passive immunization with convalescent plasma or

monoclonal antibodies seems to be ineffective [190–192],

tending to confirm that humoral responses are not primarily

responsible for the control of EBOV infection. Similarly,

recombinant type I IFN administration fails to protect non-

human primates [193]. One promising approach is to manip-

ulate the coagulation system by inhibiting the tissue factor

pathway with a factor VIIa/tissue factor inhibitor [156] or by

activating the natural anticoagulant protein C pathway with

recombinant activated protein C [194]. However, the sur-

vival benefit is limited. Another approach is to target viral

replication with small interfering RNA or antisense oligonu-

cleotides [195–198], although the latter is limited by the

need to use virus species-specific sequences, which are not

known in the early stages of an outbreak.

Conclusions

Substantial advances in our understanding of the ecology of

EBOV and MARV, as well as host–virus interactions and dis-

ease control, have been made during the past two decades.

This period was marked by the first recorded MARV out-

break in West Africa, resurgence of Ebola Sudan in Sudan

and Uganda, the discovery of two new species (Cote d’Ivoire

and Bundibugyo), and escalation of ZEBOV outbreaks in the

border region of Gabon and the Republic of Congo. Bats

have been identified as a major filovirus reservoir, and many

human outbreaks have been shown to have arisen through

the handling of infected chimpanzee and gorilla carcasses.

The 2007 ZEBOV outbreak was linked to fruit bats, although

the precise mechanism of transmission to humans was not

identified. MARV and ZEBOV induce profound suppression

of adaptive immunity, characterized by massive B-lymphocyte

and T-lymphocyte apoptosis largely mediated by the TRAIL

(TNF-related apoptosis-inducing ligand) and Fas pathways. A

recent study suggests that superantigenic activity may be

involved and numerous studies have shown that VP35 and

VP24 play an essential role in ZEBOV suppression of IFN-a/

b production.

Transparency Declaration

Conflicts of interest: nothing to declare.

8 Clinical Microbiology and Infection CMI

ª2011 The Authors

Clinical Microbiology and Infection ª2011 European Society of Clinical Microbiology and Infectious Diseases, CMI

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

21

22

23

24

25

26

27

28

29

30

31

32

33

34

35

36

37

38

39

40

41

42

43

44

45

46

47

48

49

50

References

1. Hoenen T, Groseth A, Falzarano D, Feldmann H. Ebola virus: unrav-

elling pathogenesis to combat a deadly disease. Trends Mol Med

2006; 12: 206–215.

2. Sanchez A, Geisbert TW, Feldmann H. Filoviridae: Marburg and Ebo-

la viruses. In: Knipe DM, Howley PM, eds, Fields virology. Philadelphia:

Lippincott Williams and Williams, 2007; 1409–1448.

3. Borio L, Inglesby T, Peters CJ et al. Hemorrhagic fever viruses as

biological weapons: medical and public health management. JAMA

2002; 287: 2391–2405.

4. Centers for Disease Control and Prevention. Bioterrorism agents/

diseases, 2010. Available at: http://wwwbtcdcgov/agent/agentlist-cate-

goryasp#a3 .

5. World Health Organization. Recommendations for management of

viral haemorrhagic fevers in Africa. ?????: World Health Organization,

19854

6. Bray M, Murphy FA. Filovirus research: knowledge expands to meet

a growing threat. J Infect Dis 2007; 196 (suppl. 2): S438–S443.

7. World Health Organization. Ebola haemorrhagic fever – fact sheet

revised in May 2004. Wkly Epidemiol Rec 2004; 79: 435–439.

8. Borchert M, Boelaert M, Sleurs H et al. Viewpoint: filovirus haemor-

rhagic fever outbreaks: much ado about nothing? Trop Med Int Health

2000; 5: 318–324.

9. Smith CEG, Simpson DIH, Bowen ETW. Fatal human disease from

vervet monkeys. Lancet 1967; II: 1119–1121.

10. Martini GA. Marburg agent disease in man. Trans R Soc Trop Med

Hyg 1969; 63: 295–302.

11. Martini GA, Siegert R. Marburg virus disease. New York: Springer,

1971.

12. Slenczka W, Klenk HD. Forty years of Marburg virus. J Infect Dis

2007; 196 (suppl. 2): S131–S135.

13. Smith DIH. Ebola haemorrhagic fever in Sudan, 1976. Bull World

Health Organ 1978; 56: 247–270.

14. Johnson KM. Ebola haemorrhagic fever in Zaire, 1976. Bull World

Health Organ 1978; 56: 271–293.

15. Kiley MP, Regnery RL, Johnson KM. Ebola virus: identification of vir-

ion structural proteins. J Gen Virol 1980; 49: 333–341.

16. Feldmann H, Kiley MP. Classification, structure, and replication of fil-

oviruses. Curr Top Microbiol Immunol 1999; 235: 1–21.

17. Sanchez A, Khan A, Zaki SR, Nabel GJ, Ksiazek TG, Peters CJ. Filo-

viridae: Marburg and Ebola viruses. In: Knipe DM, Howley PM, eds,

Fields virology. Philadelphia: Lippincott Williams & Wilkins, 2001;

1279–1304.

18. Kiley MP, Bowen TW, Eddy GA et al. Filoviridae: taxonomic home

for Marburg and Ebola viruses? Intervirology 1982; 18: 24–32.

19. Regnery RL, Johnson KM, Kiley MP. Virion nucleic acid of Ebola

virus. J Virol 1980; 36: 465–469.

20. Feldmann H, Muhlberger E, Randolf A et al. Marburg virus, a filo-

virus: messenger RNAs, gene order, and regulatory elements of the

replication cycle. Virus Res 1992; 24: 1–19.

21. Muhlberger E, Sanchez A, Randolf A et al. The nucleotide sequence

of the l gene of Marburg virus, a filovirus: homologies with paramyx-

oviruses and rhabdoviruses. Virology 1992; 187: 534–547.

22. Feldmann H, Klenk HD, Sanchez A. Molecular biology and evolution

of filoviruses. Arch Virol 1993; 7: 81–100.5

23. Feldmann H, Nichol ST, Klenk H-D, Peters CJ, Sanchez A. Charac-

terization of filoviruses based on difference in structure and antige-

nicity of the virion glycoprotein. Virology 1994; 199: 469–473.

24. Martini GA. Clinical syndrome. In: MGAaS R, ed. Marburg virus dis-

ease. New York: Springer-Verlag, 1971; 1–9.

25. Smith DH, Francis F, Simpson DIH. African haemorrhagic fever in

southern Sudan, 1976: the clinical manifestations. In: Pattyn SR, ed.

Ebola virus haemorrhagic fever. New York: Elsevier/North-Holland

biomedical press, 1978; 21–26.

26. Peters CJ, Khan AS. Filovirus diseases. In: Klenk HD, ed. Current top-

ics in microbiology and immunology: filovirus disease. Berlin: Springer-

Verlag, 1999; 85–95.

27. Peters CJ, LeDuc JW. An introduction to Ebola: the virus and the

disease. J Infect Dis 1999; 179 (suppl. 1): ix–xvi.

28. Bwaka MA, Bonnet M-J, Calain P et al. Ebola hemorrhagic fever in

Kiwit, Democratic Republic of the Congo: clinical observations in

103 patients. J Infect Dis 1999; 179: S1–S7.

29. Colebunders R, Tshomba A, Van Kerkhove MD et al. Marburg hem-

orrhagic fever in Durba and Watsa, Democratic Republic of the

Congo: clinical documentation, features of illness, and treatment. J

Infect Dis 2007; 196 (suppl. 2): S148–S153.

30. Emond RT, Evans B, Bowen ET, Lloyd G. A case of Ebola virus infec-

tion. Br Med J 1977; 2: 541–544.

31. Smith DH, Isaacson M, Johnson KM et al. Marburg-virus disease in

Kenya. Lancet 1982; I: 816–820.

32. Rowe AK, Bertolli J, Khan AS et al. Clinical, virologic, and immuno-

logic follow-up of convalescent Ebola hemorrhagic fever patients and

their household contacts, Kikwit, Democratic Republic of the

Congo. J Infect Dis 1999; 179: S28–S35.

33. Fisher-Hoch SP, Lloyd G, Platt GS, Simpson DIH. Hematological and

biochemical monitoring of Ebola infection in rhesus monkeys: impli-

cations for patient management. Lancet 1983; 2: 1055–1058.

34. Fisher-Hoch SP, Platt GS, Neild GH et al. Pathophysiology of shock

and hemorrhage in a fulminating viral infection (Ebola). J Infect Dis

1985; 152: 887–894.

35. Geisbert TW, Young HA, Jahrling PB, Davis KJ, Kagan E,

Hensley L. Mechanisms underlying coagulation abnormalities in Ebola

hemorrhagic fever: overexpression of tissue factor in primate

monocytes/macrophages is a key event. J Infect Dis 2003; 188: 1618–

1629.

36. Geisbert TW, Young HA, Jahrling PB et al. Pathogenesis of Ebola

hemorrhagic fever in primate models: evidence that hemorrhage is

not a direct effect of virus-induced cytolysis of endothelial cells. Am

J Pathol 2003; 163: 2371–2382.

37. Kuhn JH. Filoviruses. A compendium of 40 years of epidemiological,

clinical, and laboratory studies. Arch Virol Suppl 2008; 20: 13–360.

38. Gear JS, Cassel GA, Gear AJ et al. Outbreak of Marburg virus dis-

ease in Johannesburg. Br Med J 1975; 4: 489–493.

39. Johnson ED, Johnson BK, Silverstein D et al. Characterization of a

new Marburg virus isolated from a 1987 fatal case in Kenya. Arch

Virol Suppl 1996; 11: 101–114.

40. Bausch DG, Nichol ST, Muyembe-Tamfum JJ et al. Marburg hemor-

rhagic fever associated with multiple genetic lineages of virus. N Engl

J Med 2006; 355: 909–919.

41. Bausch DG, Borchert M, Grein T et al. Risk factors for Marburg

hemorrhagic fever, Democratic Republic of the Congo. Emerg Infect

Dis 2003; 9: 1531–1537.

42. Towner JS, Khristova ML, Sealy TK et al. Marburg virus genomics

and association with a large hemorrhagic fever outbreak in Angola.

J Virol 2006; 80: 6497–6516.

43. Towner JS, Amman BR, Sealy TK et al. Isolation of genetically

diverse Marburg viruses from Egyptian fruit bats. PLoS Pathog 2009;

5: e1000536.

44. Hartman AL, Towner JS, Nichol ST. Ebola and Marburg hemorrhagic

fever. Clin Lab Med 2010; 30: 161–177.

45. Heymann DL, Weisfeld JS, Webb PA, Johnson KM, Cairns T, Ber-

quist H. Ebola hemorrhagic fever: Tandala, 1977–1978. J Infect Dis

1980; 142: 372–376.

46. Baron RC, McCormick JB, Zubeir OA. Ebola virus disease in south-

ern Sudan: hospital dissemination and intrafamilial spread. Bull World

Health Organ 1983; 61: 997–1003.

CMI Leroy et al. Ebola and Marburg haemorrhagic fever viruses1

9

ª2011 The Authors

Clinical Microbiology and Infection ª2011 European Society of Clinical Microbiology and Infectious Diseases, CMI

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

21

22

23

24

25

26

27

28

29

30

31

32

33

34

35

36

37

38

39

40

41

42

43

44

45

46

47

48

49

50

47. Le Guenno B, Formenty P, Wyers M, Gounon P, Walker F, Boesch

C. Isolation and partial characterisation of a new strain of Ebola. Lan-

cet 1995; 345: 1271–1274.

48. Le Guenno B, Formenty P, Boesch C. Ebola virus outbreaks in Ivory

Coast and Liberia, 1994–1995. In: Klenk H-D, ed. Current topics in

microbiology and immunology: Marburg and Ebola viruses. Berlin:

Springer-Verlag, 1999; 77–84.

49. Formenty P, Hatz C, Le Guenno B, Stoll A, Rogenmoser P,

Wildmer A. Human infection due to Ebola virus, subtype Cote

d’Ivoire: clinical and biologic presentation. J Infect Dis 1999; 179:

S48–S53.

50. Khan AS, Tshioko FK, Heymann DL et al. The reemergence of Ebola

hemorrhagic fever, Democratic Republic of the Congo, 1995. J Infect

Dis 1999; 179: S76–S86.

51. Georges AJ, Leroy EM, Renaut AA et al. Ebola hemorrhagic fever

outbreaks in Gabon, 1994–1997: epidemiologic and health control

issues. J Infect Dis 1999; 179: S65–S75.

52. Amblard J, Obiang P, Edzang S, Prehaud C, Bouloy M, Le Guenno B.

Identification of the Ebola virus in Gabon in 1994. Lancet 1997; 349:

181–182.

53. Georges-Courbot M-C, Lu C-Y, Lansoud-Soukate J, Leroy E, Baize

S. Isolation and partial molecular characterisation of a strain of Ebola

virus during a recent epidemic of viral haemorrhagic fever in Gabon.

Lancet 1997; 349: 181.

54. Georges-Courbot M-C, Sanchez A, Lu C-Y et al. Isolation and phylo-

genetic characterization of Ebola viruses causing different outbreaks

in Gabon. Emerg Infect Dis 1997; 1: 59–62.

55. Leroy EM, Souquiere S, Rouquet P, Drevet D. Re-emergence of

Ebola haemorrhagic fever in Gabon. Lancet 2002; 359: 712.

56. Leroy EM, Rouquet P, Formenty P et al. Multiple Ebola virus trans-

mission events and rapid decline of central african wildlife. Science

2004; 303: 387–390.

57. Pourrut X, Kumulungui B, Wittmann T et al. The natural history of

Ebola virus in Africa. Microbes Infect 2005; 7: 1005–1014.

58. Formenty P, Leroy EM, Epelboin A et al. Detection of Ebola virus

in oral fluid specimens during outbreaks of Ebola virus hemor-

rhagic fever in the Republic of Congo. Clin Infect Dis 2006; 42: 1521–

1526.

59. Lamunu M, Lutwama JJ, Kamugisha J et al. Containing a haemorrhagic

fever epidemic: the Ebola experience in Uganda (October 2000–Jan-

uary 2001). Int J Infect Dis 2004; 8: 27–37.

60. Okware SI, Omaswa FG, Zaramba S et al. An outbreak of Ebola in

Uganda. Trop Med Int Health 2002; 7: 1068–1075.

61. World Health Organization. Ebola haemorrhagic fever in south

Sudan – update. Wkly Epidemiol Rec 2004; 79: 253.

62. Towner JS, Rollin PE, Bausch DG et al. Rapid diagnosis of Ebola

hemorrhagic fever by reverse transcription-PCR in an outbreak set-

ting and assessment of patient viral load as a predictor of outcome.

J Virol 2004; 78: 4330–4341.

63. Onyango CO, Opoka ML, Ksiazek TG et al. Laboratory diagnosis of

Ebola hemorrhagic fever during an outbreak in Yambio, Sudan, 2004.

J Infect Dis 2007; 196 (suppl. 2): S193–S198.

64. Towner JS, Sealy TK, Khristova ML et al. Newly discovered Ebola

virus associated with hemorrhagic fever outbreak in Uganda. PLoS

Pathog 2008; 4: e1000212.

65. Jahrling PB, Geisbert TW, Dalgard DW et al. Preliminary report: iso-

lation of Ebola virus from monkeys imported to USA. Lancet 1990;

335: 502–505.

66. Dalgard DW, Hardy RJ, Pearson SL et al. Combined simian hemor-

rhagic fever and Ebola virus infection in cynomolgus monkeys. Lab

Anim Sci 1992; 42: 152–157.

67. Hayes CG, Burans JP, Ksiazek TG et al. Outbreak of fatal illness

among captive macaques in the Philippines caused by an Ebola-

related filovirus. Am J Trop Med Hyg 1992; 46: 664–671.

68. Rollin PE, Williams RJ, Bressler DS et al. Ebola (subtype reston)

virus among quarantined nonhuman primates recently imported

from the Philippines to the United States. J Infect Dis 1999; 179:

S108–S114.

69. Miranda MEG, White ME, Dayrit MM, Hayes CG, Ksiazek TG, Bu-

rans JP. Seroepidemiological study of filovirus related to Ebola in the

Philippines. Lancet 1991; 337: 425–426.

70. Miranda ME, Ksiazek TG, Retuya TJ et al. Epidemiology of Ebola

(subtype reston) virus in the Philippines, 1996. J Infect Dis 1999; 179:

S115–S119.

71. Barrette RW, Metwally SA, Rowland JM et al. Discovery of swine as

a host for the reston Ebola virus. Science 2009; 325: 204–206.

72. Huang Y, Xu L, Sun Y, Nabel GJ. The assembly of Ebola virus

nucleocapsid requires virion-associated proteins 35 and 24 and post-

translational modification of nucleoprotein. Mol Cell 2002; 10: 307–

316.

73. Muhlberger E, Weik M, Volchkov VE, Klenk H-D, Becker S. Compari-

son of the transcription and replication strategies of Marburg and

Ebola virus by using artificial replication systems. J Virol 1999; 73: ???–

???. 6

74. Volchkov VE, Volchkova VA, Muhlberger E et al. Recovery of infec-

tious Ebola virus from complementary DNA: RNA editing of the gp

gene and viral cytotoxicity. Science 2001; 291: 1965–1969.

75. Volchkov VE, Chepurnov AA, Volchkova VA, Ternovoj VA, Klenk

HD. Molecular characterization of guinea pig-adapted variants of

Ebola virus. Virology 2000; 277: 147–155.

76. Noda T, Sagara H, Suzuki E, Takada A, Kida H, Kawaoka Y. Ebola

virus vp40 drives the formation of virus-like filamentous particles

along with gp. J Virol 2002; 76: 4855–4865.

77. Han Z, Boshra H, Sunyer JO, Zwiers SH, Paragas J, Harty RN. Bio-

chemical and functional characterization of the Ebola virus vp24 pro-

tein: implications for a role in virus assembly and budding. J Virol

2003; 77: 1793–1800.

78. Watanabe S, Watanabe T, Noda T et al. Production of novel Ebola

virus-like particles from cDNAs: an alternative to Ebola virus gener-

ation by reverse genetics. J Virol 2004; 78: 999–1005.

79. Bamberg S, Kolesnikova L, Moller P, Klenk HD, Becker S. Vp24 of

Marburg virus influences formation of infectious particles. J Virol

2005; 79: 13421–13433.

80. Noda T, Watanabe S, Sagara H, Kawaoka Y. Mapping of the vp40-

binding regions of the nucleoprotein of Ebola virus. J Virol 2007; 81:

3554–3562.

81. Johnson RF, McCarthy SE, Godlewski PJ, Harty RN. Ebola virus

vp35-vp40 interaction is sufficient for packaging 3e-5e minigenome

RNA into virus-like particles. J Virol 2006; 80: 5135–5144.

82. Ruigrok RW, Schoehn G, Dessen A et al. Structural characterization

and membrane binding properties of the matrix protein vp40 of

Ebola virus. J Mol Biol 2000; 300: 103–112.

83. Sanchez A, Trappier SG, Mahy BWJ, Peters CJ, Nichol ST. The vir-

ion glycoproteins of Ebola viruses are encoded in two reading

frames and are expressed through transcriptional editing. Proc Natl

Acad Sci USA 1996; 93: 3602–3607.

84. Volchkov VE, Feldmann H, Volchkova VA, Klenk H-D. Processing of

the Ebola virus glycoprotein by the proprotein convertase furin. Proc

Natl Acad Sci USA 1998; 95: 5762–5767.

85. Volchkov VE, Volchkova VA, Slenczka W, Klenk H-D, Feldmann H.

Release of viral glycoproteins during Ebola virus infection. Virology

1998; 245: 110–119.

86. Volchkov VE, Blinov VM, Netesov SV. The envelope glycoprotein of

Ebola virus contains an immunosuppressive-like domain similar to

oncogenic retroviruses. FEBS Lett 1992; 305: 181–184.

87. Wool-Lewis RJ, Bates P. Characterization of Ebola virus entry by

using pseudotyped viruses: identification of receptor-deficient cell

lines. J Virol 1998; 72: 3155–3160.

10 Clinical Microbiology and Infection CMI

ª2011 The Authors

Clinical Microbiology and Infection ª2011 European Society of Clinical Microbiology and Infectious Diseases, CMI

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

21

22

23

24

25

26

27

28

29

30

31

32

33

34

35

36

37

38

39

40

41

42

43

44

45

46

47

48

49

50

88. Yang Z-Y, Delgado R, Xu L et al. Distinct cellular interactions of

secreted and transmenbrane Ebola virus glycoproteins. Science 1998;

279: 1034–1037.

89. Chepurnov AA, Tuzova MN, Ternovoy VA, Chernukhin IV.

Suppressive effect of Ebola virus on T cell proliferation in vitro is

provided by a 125-kDa gp viral protein. Immunol Lett 1999; 68: 257–

261.

90. Yang ZY, Duckers HJ, Sullivan NJ, Sanchez A, Nabel EG, Nabel GJ.

Identification of the Ebola virus glycoprotein as the main viral deter-

minant of vascular cell cytotoxicity and injury. Nat Med 2000; 6:

886–889.

91. Chan SY, Speck RF, Ma MC, Goldsmith MA. Distinct mechanisms of

entry by envelope glycoproteins of Marburg and Ebola (Zaire)

viruses. J Virol 2000; 74: 4933–4937.

92. Chan SY, Empig CJ, Welte FJ et al. Folate receptor-alpha is a cofac-

tor for cellular entry by Marburg and Ebola viruses. Cell 2001; 106:

117–126.

93. Feldmann H, Volchkov VE, Volchkova VA, Stroher U, Klenk HD.

Biosynthesis and role of filoviral glycoproteins. J Gen Virol 2001; 82:

2839–2848.

94. Yaddanapudi K, Palacios G, Towner JS et al. Implication of a retrovi-

rus-like glycoprotein peptide in the immunopathogenesis of Ebola

and Marburg viruses. FASEB J 2006; 20: 2519–2530.

95. Volchkova VA, Feldmann H, Klenk H-D, Volchkov VE. The non-

structural small glycoprotein sgp of Ebola virus is secreted as an

antiparallel-orientated homodimer. Virology 1998; 250: 408–414.

96. Dolnik O, Volchkova V, Garten W et al. Ectodomain shedding of the

glycoprotein gp of Ebola virus. EMBO J 2004; 23: 2175–2184.

97. Basler CF, Mikulasova A, Martinez-Sobrido L et al. The Ebola virus

vp35 protein inhibits activation of interferon regulatory factor 3. J

Virol 2003; 77: 7945–7956.

98. Hartman AL, Towner JS, Nichol ST. A C-terminal basic amino acid

motif of Zaire Ebola virus vp35 is essential for type I interferon

antagonism and displays high identity with the RNA-binding domain

of another interferon antagonist, the ns1 protein of influenza A

virus. Virology 2004; 328: 177–184.

99. Cardenas WB, Loo YM, Gale M Jr et al. Ebola virus vp35 protein

binds double-stranded RNA and inhibits alpha/beta interferon pro-

duction induced by RIG-I signaling. J Virol 2006; 80: 5168–5178.

100. Hartman AL, Bird BH, Towner JS, Antoniadou ZA, Zaki SR, Nichol

ST. Inhibition of IRF-3 activation by vp35 is critical for the high level

of virulence of Ebola virus. J Virol 2008; 82: 2699–2704.

101. Prins KC, Cardenas WB, Basler CF. Ebola virus protein vp35 impairs

the function of interferon regulatory factor-activating kinases IKKep-

silon and TBK-1. J Virol 2009; 83: 3069–3077.

102. Chang TH, Kubota T, Matsuoka M et al. Ebola Zaire virus blocks

type I interferon production by exploiting the host Sumo modifica-

tion machinery. PLoS Pathog 2009; 5: e1000493.

103. Suzuki Y, Gojobori T. The origin and evolution of Ebola and Mar-

burg viruses. Mol Biol Evol 1997; 14: 800–806.

104. Rodriguez LL, De Roo A, Guimard Y et al. Persistence and genetic

stability of Ebola virus during the outbreak in Kikwit, Democratic

Republic of the Congo, 1995. J Infect Dis 1999; 179: S170–S176.

105. Leroy EM, Baize S, Lansoud-Soukate J, Mavoungou E, Apetrei C.

Sequence analysis of gp, np, vp40 and vp24 genes of Ebola virus from

deceased, survival and asymptomatic infected individuals during 1996

outbreak in Gabon. Comparative studies and phylogenetic character-

ization. J Gen Virol 2002; 83: 67–73.

106. Sanchez A, Rollin PE. Complete genome sequence of an Ebola virus

(Sudan species) responsible for a 2000 outbreak of human disease in

Uganda. Virus Res 2005; 113: 16–25.

107. Wittmann TJ, Biek R, Hassanin A et al. Isolates of Zaire Ebola virus

from wild apes reveal genetic lineage and recombinants. Proc Natl

Acad Sci U S A 2007; 104: 17123–17127.

108. Pinzon JE, Wilson JM, Tucker CJ, Arthur R, Jahrling PB, Formenty P.

Trigger events: enviroclimatic coupling of Ebola hemorrhagic fever

outbreaks. Am J Trop Med Hyg 2004; 71: 664–674.

109. Peterson AT, Bauer JT, Mills JN. Ecologic and geographic distribution

of filovirus disease. Emerg Infect Dis 2004; 10: 40–47.

110. Peterson AT, Carroll DS, Mills JN, Johnson AM. Potential mamma-

lian filovirus reservoirs. Emerg Infect Dis 2004; 10: 2073–2081.

111. Peterson AT, Lash RR, Carroll DS, Johnson KM. Geographic poten-

tial for outbreaks of Marburg hemorrhagic fever. Am J Trop Med Hyg

2006; 75: 9–15.

112. Becquart P, Wauquier N, Mahlakoiv T et al. High prevalence of both

humoral and cellular immunity to Zaire Ebola virus among rural pop-

ulations in Gabon. PLoS ONE 2010; 5: e9126.

113. Ivanoff B, Duquesnoy P, Languillat G et al. Haemorrhagic fever in

Gabon. I. Incidence of Lassa, Ebola and Marburg viruses in Haut-Og-

ooue. Trans R Soc Trop Med Hyg 1982; 76: 719–720.

114. Bouree P, Bergmann JF. Ebola virus infection in man: a serological

and epidemiological survey in the Cameroons. Am J Trop Med Hyg

1983; 32: 1465–1466.

115. Gonzalez JP, Josse R, Johnson ED et al. Antibody prevalence against

haemorrhagic fever viruses in randomized representative Central

African populations. Res Virol 1989; 140: 319–331.

116. Johnson BK, Wambui C, Ocheng D et al. Seasonal variation in anti-

bodies against Ebola virus in Kenyan fever patients. Lancet 1986; 1:

1160.

117. Johnson ED, Gonzalez JP, Georges AJ. Filovirus activity among

selected ethnic groups inhabiting the tropical forest of Equatorial

Africa. Trans R Soc Trop Med Hyg 1993; 87: 536–538.

118. Busico KM, Marshall KL, Ksiazek TG et al. Prevalence of IgG antibod-

ies to Ebola virus in individuals during an Ebola outbreak, Democratic

Republic of the Congo, 1995. J Infect Dis 1999; 179: S102–S107.

119. Gonzalez JP, Nakoune E, Slenczka W, Vidal P, Morvan JM. Ebola and

Marburg virus antibody prevalence in selected populations of the

Central African Republic. Microbes Infect 2000; 2: 39–44.

120. Leroy EM, Baize S, Volchkov VE et al. Human asymptomatic Ebola

infection and strong inflammatory response. Lancet 2000; 355: 2210–

2215.

121. Huijbregts B, De Wachter P, Ndong Obiang S, Akou Ella M. Ebola

and the decline of gorilla Gorilla gorilla and chimpanzee Pan troglodytes

populations in Minkebe forest, northeastern Gabon. Oryx 2003; 37:

437–443.

122. Walsh PD, Abernethy KA, Bermejo M et al. Catastrophic ape

decline in western Equatorial Africa. Nature 2003; 422: 611–614.

123. Rouquet P, Froment J-M, Bermejo M et al. Wild animal mortality

monitoring and human Ebola outbreaks, Gabon and Republic of

Congo, 2001–2003. Emerg Infect Dis 2005; 11: 283–290.

124. Bermejo M, Rodriguez-Teijeiro JD, Illera G, Barroso A, Vila C,

Walsh PD. Ebola outbreak killed 5000 gorillas. Science 2006; 314:

1564.

125. Caillaud D, Levrero F, Cristescu R et al. Gorilla susceptibility to

Ebola virus: the cost of sociality. Curr Biol 2006; 16: R489–R491.

126. Formenty P, Boesch C, Wyers M et al. Ebola virus outbreak among

wild chimpanzees living in a rain forest of Cote d’Ivoire. J Infect Dis

1999; 179 (suppl. 1): S120–S126.

127. Leroy EM, Kumulungui B, Pourrut X et al. Fruit bats as reservoirs of

Ebola virus. Nature 2005; 438: 575–576.

128. Monath TP. Ecology of Marburg and Ebola viruses: speculations

and directions for the future research. J Infect Dis 1999; 179: S127–

S138.

129. Leroy EM, Telfer P, Kumulungui B et al. A serological survey of Ebo-

la virus infection in Central African nonhuman primates. J Infect Dis

2004; 190: 1895–1899.

130. Feldmann H, Wahl-Jensen V, Jones SM, Stroher U. Ebola virus ecol-

ogy: a continuing mystery. Trends Microbiol 2004; 12: 433–437.

CMI Leroy et al. Ebola and Marburg haemorrhagic fever viruses1

11

ª2011 The Authors

Clinical Microbiology and Infection ª2011 European Society of Clinical Microbiology and Infectious Diseases, CMI

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

21

22

23

24

25

26

27

28

29

30

31

32

33

34

35

36

37

38

39

40

41

42

43

44

45

46

47

48

49

50

131. Arata AA, Johnson B. Approaches towards studies on potential res-

ervoirs of viral haemorrhagic fever in southern Sudan (1977). In: Pat-

tyn SR, ed. Ebola virus haemorrhagic fever. Amsterdam: Elsevier/

Netherland biomedical, 1978; 191–202.

132. Turell MJ, Bressler DS, Rossi CA. Lack of virus replication in arthro-

pods after intrathoracic inoculation of Ebola reston virus. Am J Trop

Med Hyg 1996; 55: 89–90.

133. Swanepoel R, Leman PA, Burt FJ. Experimental inoculation of plants

and animals with Ebola virus. Emerg Infect Dis 1996; 2: 321–325.

134. Breman JG, Johnson KM, van der Groen G et al. A search for Ebola

virus in animals in the Democratic Republic of the Congo and Cam-

eroon: ecologic, virologic, and serologic surveys, 1979–1980. J Infect

Dis 1999; 179: S139–S147.

135. Reiter P, Turell M, Coleman R et al. Field investigations of an out-

break of Ebola hemorrhagic fever, Kikwit, Democratic Republic of the

Congo, 1995: arthropod studies. J Infect Dis 1999; 179: S148–S154.

136. Leirs H, Mills JN, Krebs JW et al. Search for the Ebola virus reser-

voir in Kikwit, Democratic Republic of the Congo: reflections on a

vertebrate collection. J Infect Dis 1999; 179: S155–S163.

137. Formenty P, Jahrling P, Rossi C et al. Search for the Ebola virus res-

ervoir in Taı forest, Cote d’Ivoire: 1996–1997, preliminary results.

XIth International Congress of Virology. Sydney, Australia, 1999.

138. Morvan JM, Deubel V, Gounon P et al. Identification of Ebola virus

sequences present as RNA or DNA in organs of terrestrial small

mammals of the Central African Republic. Microbes Infect 1999; 1:

1193–1201.

139. Pourrut X, Delicat A, Rollin PE, Ksiazek TG, Gonzalez JP, Leroy EM.

Spatial and temporal patterns of Zaire Ebola virus antibody preva-

lence in the possible reservoir bat species. J Infect Dis 2007; 196

(suppl. 2): S176–S183.

140. Towner JS, Pourrut X, Albarino CG et al. Marburg virus infection

detected in a common African bat. PLoS ONE 2007; 2: e764.

141. Swanepoel R, Smit SB, Rollin PE et al. Studies of reservoir hosts for

Marburg virus. Emerg Infect Dis 2007; 13: 1847–1851.

142. Leroy EM, Epelboin A, Mondonge V et al. Human Ebola outbreak

resulting from direct exposure to fruit bats in Luebo, Democratic

Republic of Congo, 2007. Vector Borne Zoonotic Dis 2009; 6: 723–

728.

143. Mahanty S, Hutchinson K, Agarwal S, McRae M, Rollin PE, Pulendran

B. Cutting edge: impairment of dendritic cells and adaptive immunity

by Ebola and Lassa viruses. J Immunol 2003; 170: 2797–2801.

144. Stroher U, West E, Bugany H, Klenk H-D, Schnittler H-J, Feldmann

H. Infection and activation of monocytes by Marburg and Ebola

viruses. J Virol 2001; 75: 11025–11033.

145. Geisbert TW, Hensley L, Larsen T et al. Pathogenesis of Ebola hem-

orrhagic fever in cynomolgus macaques: evidence that dendritic cells

are early and sustained targets of infection. Am J Pathol 2003; 163:

2347–2370.

146. Ryabchikova EI, Kolesnikova LV, Luchko SV. An analysis of features

of pathogenesis in two animal models of Ebola virus infection. J Infect

Dis 1999; 179: S199–S202.

147. Baskerville A, Fisher-Hoch SP, Neild GH, Dowsett AB. Ultrastruc-

ture pathology of experimental Ebola haemorrhagic fever virus infec-

tion. J Pathol 1985; 147: 199–209.

148. Gupta M, Mahanty S, Ahmed R, Rollin PE. Monocyte-derived human

macrophage and peripheral blood mononuclear cells infected with

Ebola virus secrete MIP-1a and TNF-a and inhibit poly-IC-induced

IFN-a in vitro. Virology 2001; 284: 20–25.

149. Baize S, Leroy EM, Georges AJ et al. Inflammatory responses in Ebo-

la virus-infected patients. Clin Exp Immunol 2001; 128: 163–168.

150. Wauquier N, Becquart P, Padilla C, Baize S, Leroy EM. Human fatal

Zaire Ebola virus infection is associated with an aberrant innate

immunity and massive lymphocyte apoptosis. PLoS Negl Trop Dis

2010; 4: e837.

151. Villinger F, Rollin PE, Brar SS et al. Markedly elevated levels of inter-

feron (IFN)-gamma, IFN-alpha, interleukin (IL)-2, IL-10, and tumor

necrosis factor-a associated with fatal Ebola virus infection. J Infect

Dis 1999; 179: S188–S191.

152. Feldmann H, Bugany H, Mahner F, Klenk HD, Drenckhahn D,

Schnittler HJ. Filovirus-induced endothelial leakage triggered by

infected monocytes/macrophages. J Clin Invest 1996; 70: 2208–

2214.

153. Schnittler HJ, Feldmann H. Molecular pathogenesis of filovirus infec-

tions: role of macrophages and endothelial cells. Curr Top Microbiol

Immunol 1999; 235: 175–204.

154. Hensley L, Young HA, Jahrling PB, Geisbert TW. Proinflammatory

response during Ebola virus infection of primate models: possible

involvement of the tumor necrosis factor receptor superfamily.

Immunol Lett 2002; 80: 169–179.

155. Gerszten RE, Garcia-Zepeda EA, Lim Y-C et al. MCP-1 and IL-8 trig-

ger firm adhesion of monocytes to vascular endothelium under flow

conditions. Nature 1999; 398: 718–723.

156. Geisbert TW, Hensley L, Jahrling PB et al. Treatment of Ebola virus

infection with a recombinant inhibitor of factor VII/tissue factor: a

study in rhesus monkeys. Lancet 2003; 362: 1953–1958.

157. Ruf W. Emerging roles of tissue factor in viral hemorrhagic fever.

Trends Immunol 2004; 25: 461–464.

158. Baize S, Leroy EM, Georges-Courbot M-C et al. Defective humoral

responses and extensive intravascular apoptosis are associated with

fatal outcome in Ebola virus-infected patients. Nat Med 1999; 5:

423–426.

159. Baize S, Leroy EM, Mavoungou E, Fisher-Hoch SP. Apoptosis in fatal

Ebola infection. Does the virus toll the bell for the immune system?

Apoptosis 2000; 5: 5–7.

160. Geisbert TW, Hensley LE, Gibb TR, Steele KE, Jaax NK, Jahrling PB.

Apoptosis induced in vitro and in vivo during infection by Ebola and

Marburg viruses. Lab Invest 2000; 80: 171–186.

161. Reed DS, Hensley LE, Geisbert JB, Jahrling PB, Geisbert TW. Deple-

tion of peripheral blood T lymphocytes and NK cells during the

course of Ebola hemorrhagic fever in cynomolgus macaques. Viral

Immunol 2004; 17: 390–400.

162. Leroy EM, Becquart P, Wauquier N, Baize S. Evidence for Ebola

virus superantigen activity. J Virol 2011; ???: ???–???. 7

163. Bradfute SB, Swanson PE, Smith MA et al. Mechanisms and conse-

quences of ebolavirus-induced lymphocyte apoptosis. J Immunol

2010; 184: 327–335.

164. Parrino J, Hotchkiss RS, Bray M. Prevention of immune cell apopto-

sis as potential therapeutic strategy for severe infections. Emerg

Infect Dis 2007; 13: 191–198.

165. Ksiazek TG, Rollin PE, Williams AJ et al. Clinical virology of Ebola

hemorrhagic fever (EHF): virus, virus antigen, and IgG and IgM anti-

body findings among EHF patients in Kikwit, Democratic Republic of

the Congo, 1995. J Infect Dis 1999; 179: S177–S187. 8

166. Bosio CM, Aman MJ, Grogan C et al. Ebola and Marburg viruses rep-

licate in monocyte-derived dendritic cells without inducing the pro-

duction of cytokines and full maturation. J Infect Dis 2003; 188:

1630–1638.

167. Jin H, Yan Z, Prabhakar BS et al. The vp35 protein of Ebola virus

impairs dendritic cell maturation induced by virus and lipopolysac-

charide. J Gen Virol 2010; 91: 352–361.

168. Basler CF, Wang X, Muhlberger E et al. The Ebola virus vp35 pro-

tein functions as a type I IFN antagonist. Proc Natl Acad Sci U S A

2000; 97: 12289–12294.

169. Feng Z, Cerveny M, Yan Z, He B. The vp35 protein of Ebola virus

inhibits the antiviral effect mediated by double-stranded RNA-depen-

dent protein kinase PKR. J Virol 2007; 81: 182–192.

170. Fabozzi G, Nabel CS, Dolan MA, Sullivan NJ. Ebolavirus proteins

suppress the effects of small interfering RNA by direct interaction

12 Clinical Microbiology and Infection CMI

ª2011 The Authors

Clinical Microbiology and Infection ª2011 European Society of Clinical Microbiology and Infectious Diseases, CMI

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

21

22

23

24

25

26

27

28

29

30

31

32

33

34

35

36

37

38

39

40

41

42

43

44

45

46

47

48

49

50

with the mammalian RNA interference pathway. J Virol 2011; 85:

2512–2523.

171. Haasnoot J, de Vries W, Geutjes EJ, Prins M, de Haan P, Berkhout

B. The Ebola virus vp35 protein is a suppressor of RNA silencing.

PLoS Pathog 2007; 3: e86.

172. Reid SP, Leung LW, Hartman AL et al. Ebola virus vp24 binds kary-

opherin alpha1 and blocks STAT1 nuclear accumulation. J Virol 2006;

80: 5156–5167.

173. Reid SP, Valmas C, Martinez O, Sanchez A, Basler CF. Ebola virus

vp24 proteins inhibit the interaction of np1-1 subfamily karyopherin

alpha proteins with activated STAT1. J Virol 2007; 81: 13469–13477.

174. Hartman AL, Ling L, Nichol ST, Hibberd ML. Whole-genome

expression profiling reveals that inhibition of host innate immune

response pathways by Ebola virus can be reversed by a single amino

acid change in the vp35 protein. J Virol 2008; 82: 5348–5358.

175. Prins KC, Delpeut S, Leung DW et al. Mutations abrogating vp35

interaction with double-stranded RNA render Ebola virus avirulent

in guinea pigs. J Virol 2010; 84: 3004–3015.

176. Wauquier N, Becquart P, Gasquet C, Leroy EM. Immunoglobulin G

in Ebola outbreak survivors, Gabon. Emerg Infect Dis 2009; 15:

1136–1137.

177. Leroy EM, Baize S, Debre P, Lansoud-Soukate J, Mavoungou E. Early

immune responses accompanying human asymptomatic Ebola infec-

tions. Clin Exp Immunol 2001; 124: 453–460.

178. Wauquier N, Padilla C, Becquart P, Leroy E, Vieillard V. Association

of kir2ds1 and kir2ds3 with fatal outcome in Ebola virus infection.

Immunogenetics 2010; 62: 767–771.

179. Bukreyev A, Rollin PE, Tate MK et al. Successful topical respiratory

tract immunization of primates against Ebola virus. J Virol 2007; 81:

6379–6388.

180. Geisbert TW, Geisbert JB, Leung A et al. Single-injection vaccine

protects nonhuman primates against infection with Marburg virus

and three species of Ebola virus. J Virol 2009; 83: 7296–7304.

181. Jones SM, Feldmann H, Stroher U et al. Live attenuated recombinant

vaccine protects nonhuman primates against Ebola and Marburg

viruses. Nat Med 2005; 11: 786–790.

182. Sullivan NJ, Geisbert TW, Geisbert JB et al. Accelerated vaccination

for Ebola virus haemorrhagic fever in non-human primates. Nature

2003; 424: 681–684.

183. Sullivan NJ, Geisbert TW, Geisbert JB et al. Immune protection of

nonhuman primates against Ebola virus with single low-dose adenovi-

rus vectors encoding modified gps. PLoS Med 2006; 3: e177.

184. Warfield KL, Swenson DL, Olinger GG, Kalina WV, Aman MJ, Bavari

S. Ebola virus-like particle-based vaccine protects nonhuman pri-

mates against lethal Ebola virus challenge. J Infect Dis 2007; 196 (sup-

pl. 2): S430–S437.

185. Pratt WD, Wang D, Nichols DK et al. Protection of nonhuman pri-

mates against two species of Ebola virus infection with a single com-

plex adenovirus vector. Clin Vaccine Immunol 2010; 17: 572–581.

186. Swenson DL, Wang D, Luo M et al. Vaccine to confer to nonhuman

primates complete protection against multistrain Ebola and Marburg

virus infections. Clin Vaccine Immunol 2008; 15: 460–467.

187. Hensley LE, Mulangu S, Asiedu C et al. Demonstration of cross-pro-

tective vaccine immunity against an emerging pathogenic Ebolavirus

species. PLoS Pathog 2010; 6: e1000904.

188. Geisbert TW, Daddario-DiCaprio KM, Williams KJ et al. Recombi-

nant vesicular stomatitis virus vector mediates postexposure protec-

tion against Sudan Ebola hemorrhagic fever in nonhuman primates. J

Virol 2008; 82: 5664–5668.

189. Feldmann H, Jones SM, Daddario-DiCaprio KM et al. Effective post-

exposure treatment of Ebola infection. PLoS Pathog 2007; 3: e2.

190. Geisbert TW, Hensley L, Geisbert JB, Jahrling PB. Evidence against

an important role for infectivity-enhancing antibodies in Ebola virus

infections. Virology 2002; 293: 15–19.

191. Jahrling PB, Geisbert JB, Swearengen JR, Larsen T, Geisbert TW.

Ebola hemorrhagic fever: evaluation of passive immunotherapy in

nonhuman primates. J Infect Dis 2007; 196 (suppl. 2): S400–S403.

192. Mupapa K, Massamba M, Kibadi K et al. Treatment of Ebola hemor-

rhagic fever with blood transfusions from convalescent patients. J

Infect Dis 1999; 179: S18–S23.

193. Jahrling PB, Geisbert TW, Geisbert JB et al. Evaluation of immune

globulin and recombinant interferon-a2b for treatment of experi-

mental Ebola virus infections. J Infect Dis 1999; 179: S224–S234.

194. Hensley LE, Stevens EL, Yan SB et al. Recombinant human activated

protein c for the postexposure treatment of Ebola hemorrhagic

fever. J Infect Dis 2007; 196 (suppl. 2): S390–S399.

195. Geisbert TW, Lee AC, Robbins M et al. Postexposure protection of

non-human primates against a lethal Ebola virus challenge with RNA

interference: a proof-of-concept study. Lancet 2010; 375: 1896–1905.

196. Warfield KL, Swenson DL, Olinger GG et al. Gene-specific counter-

measures against Ebola virus based on antisense phosphorodiamidate

morpholino oligomers. PLoS Pathog 2006; 2: e1.

197. Warren TK, Warfield KL, Wells J et al. Antiviral activity of a small-

molecule inhibitor of filovirus infection. Antimicrob Agents Chemother

2010; 54: 2152–2159.

198. Warren TK, Warfield KL, Wells J et al. Advanced antisense thera-

pies for postexposure protection against lethal filovirus infections.

Nat Med 2010; 16: 991–994.

CMI Leroy et al. Ebola and Marburg haemorrhagic fever viruses1

13

ª2011 The Authors

Clinical Microbiology and Infection ª2011 European Society of Clinical Microbiology and Infectious Diseases, CMI

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

21

22

23

24

25

26

27

28

29

30

31

32

33

34

35

36

37

38

39

40

41

42

43

44

45

46

47

48

49

50

Author Query Form

Journal: CLM

Article: 3535

Dear Author,

During the copy-editing of your paper, the following queries arose. Please respond to these by marking up your

proofs with the necessary changes/additions. Please write your answers on the query sheet if there is insufficient

space on the page proofs. Please write clearly and follow the conventions shown on the attached corrections

sheet. If returning the proof by fax do not write too close to the paper’s edge. Please remember that illegible

mark-ups may delay publication.

Many thanks for your assistance.

Query

reference

Query Remarks

Q1 AUTHOR: A running head short title was not supplied; please

check if this one is suitable and, if not, please supply a short title

of up to 40 characters that can be used instead.

Q2 AUTHOR: Please confirm that this is the correct expansion of

IFAT

Q3 AUTHOR: Please check this website address and confirm that it

is correct. (Please note that it is the responsibility of the

author(s) to ensure that all URLs given in this article are correct

and useable.)

Q4 AUTHOR: Please provide the address for reference [5].

Q5 AUTHOR: Please check the issue for reference [22].

Q6 AUTHOR: Please provide the page range for reference [73].

Q7 AUTHOR: Please provide the volume number, page range for ref-

erence [162].

Q8 AUTHOR: Please check authors name for reference [165].

Q9 AUTHOR: Please confirm that permission has been granted to

reproduce this figure

Q10 AUTHOR: Figure 2 has been saved at a low resolution of 244 dpi.

Please resupply at 300 dpi. Check required artwork specifications

at http://authorservices.wiley.com/submit_illust.asp?site=1

Q11 AUTHOR: Figure 3 has been saved at a low resolution of 214 dpi.

Please resupply at 300 dpi. Check required artwork specifications

at http://authorservices.wiley.com/submit_illust.asp?site=1

MARKED PROOF

Please correct and return this set

Instruction to printer

Leave unchanged under matter to remain

through single character, rule or underline

New matter followed by

or

or

or

or

or

or

or

or

or

and/or

and/or

e.g.

e.g.

under character

over character

new character

new characters

through all characters to be deleted

through letter or

through characters

under matter to be changed

under matter to be changed

under matter to be changed

under matter to be changed

under matter to be changed

Encircle matter to be changed

(As above)

(As above)

(As above)

(As above)

(As above)

(As above)

(As above)

(As above)

linking characters

through character or

where required

between characters or

words affected

through character or

where required

or

indicated in the margin

Delete

Substitute character or

substitute part of one or

more word(s)Change to italics

Change to capitals

Change to small capitals

Change to bold type

Change to bold italic

Change to lower case

Change italic to upright type

Change bold to non-bold type

Insert ‘superior’ character

Insert ‘inferior’ character

Insert full stop

Insert comma

Insert single quotation marks

Insert double quotation marks

Insert hyphen

Start new paragraph

No new paragraph

Transpose

Close up

Insert or substitute space

between characters or words

Reduce space betweencharacters or words

Insert in text the matter

Textual mark Marginal mark

Please use the proof correction marks shown below for all alterations and corrections. If you

in dark ink and are made well within the page margins.

wish to return your proof by fax you should ensure that all amendments are written clearly