Virus Activation by Host Proteinases. A Pivotal ... - Wiley Online Library

13 downloads 3544 Views 2MB Size Report
Viral tissue tropism in a susceptible host is often ... (LZ), occurring immediately after the fusion domain ... domain (CRD) consisting of 8 cysteine residues in the.
Microbiol. Immunol., 39(1), 1-9, 1995

Minireview

Virus Activation by Host Proteinases. Role in the Spread of Infection, Tissue Tropism and Pathogenicity Yoshiyuki

A Pivotal

Nagai

Department of Viral Infection, Institute of Medical Science, University of Tokyo, Shirokanedai 4-6-1, Minato-ku, Tokyo 108, Japan Received

September

Key words:

26, 1994

Cleavage-activation,

Envelope

glycoprotein,

Cellular

Viral tissue tropism in a susceptible host is often determined by virus-receptor interactions. Nevertheless, closely related viruses utilizing the same receptor molecules can display striking differences in tropism, or a virus can cause a localized infection despite the widespread occurrence of the receptor. Virus-receptor interactions cannot explain these events. Instead, the cellular proteinases required for the activation of viral fusion glycoproteins play a pivotal role. This proteinase-dependent mechanism of viral tropism was first demonstrated with Newcastle disease virus (NDV), an avian paramyxovirus, which uses ubiquitous sialic acids in complex sugars as the receptor but shows striking strain-dependent differences in tropism and virulence (35), and this mechanism is now applicable to many other virus-host systems (32, 33). of

The fusion (F) glycoprotein of NDV and other paramyxoviruses induces fusion between the viral envelope and host cell membrane, and hence enables the viral genome to enter the target cells. An essential step in F protein biosynthesis is the posttranslational endoproteolysis of the inactive precursor F0 glycoprotein by host cell proteinases (14, 35, 43). The precursor F0 is synthesized as a single chain molecule (Fig. Abbreviations:

FX,

immunodeficiency PC, proprotein respiratory

blood

virus convertase;

syncytial

clotting

factor

X; HIV-1,

type 1; NDV, Newcastle

virus;

PIV3,

parainfluenza

VAP, virus-activating

disease

Viral

pathogenicity

1). The signal peptide (SP) at the amino terminus directs the nascent polypeptide into the secretory pathway and is then removed from the nascent chain. A hydrophobic region near the carboxyl terminus (TMD) stops the translocation process and anchors the molecule to the lipid bilayer. The F0 is cleaved at the carboxyl side of a particular arginine to yield the twochain molecule with the aminoterminal F2 and the carboxy-terminalF1subunits, which remain bound together by a disulfide bond. The cleavage results in exposure of a long (about 25 amino acids) hydrophobic fusion-inducing domain (FID) at the amino terminus of the transmembrane F1 subunit (7). Thus, the overall structural features of fusion glycoprotein include three hydrophobic domains: the signal peptide, fusion domain and membrane anchor domain.Three other important structural domains are predicted (Fig. 1). Two of these are the heptad repeat (HR) and the leucine zipper (LZ), occurring immediately after the fusion domain and before the transmembrane domain, respectively (5, 6), and are thought to be important for oligomerization to form a tetramer, the mature form of the glycoprotein spike. The third one is the cysteine-rich domain (CRD) consisting of 8 cysteine residues in the middle of the F1 subunit. Their precise conservation in the primary sequence among paramyxovirus F proteins and the viral behavior to specificmonoclonal antibodies have suggested functional and antigenic importance of this domain (38, 52). We have recently shown with the mature F glycoprotein of Sendai virus, a murine paramyxovirus, that the cysteine residues all participate in disulfide-bridges, forming a bunched structure including two tandem loops, and we also were able to identify the cysteine residues for the intermolecular bridge between the F2 and F, subunits

1. Introduction

2. Structure, Function and Cleavage-Activation Paramyxovirus Fusion Glycoprotein

proteinase,

human virus;

virus

3; RSV , proteinase .

1

2

Y. NAGAI

3. Cleavage-Site-Motif Determines Availability of Activating Proteinases, Hence Viral Tissue Tropism and Pathogenicity

place in the Golgi apparatus (31, 36), more specifically in the trans Golgi network, while the latter is an extracellular event, as described later. The amino acid sequence at the cleavage site appears to be the most important determinant for the different cleavability of the glycoproteins. The readily cleavable Fos usually have a pair or a cluster of basic residues at this position whereas a single arginine is found with these of limited cleavability(Table 1) (53, 54). Obviously, the coincidence among cleavage site motifs, tissue specificity of proteinases for activation and the mode of virus spreading is also seen for other paramyxoviruses (Table 1). The Sendai virus receptor is ubiquitous sialic acids; however, the virus targets only the respiratory tract. Apparently, a specific trypsin-like proteinase recognizes the single-arginine motif in F0of Sendai virus, facilitates cleavage and allows spread of the virus, ultimately to cause pneumonia (49). The absence of significant virus growth in other organs is probably due to the absence of specific virus-activatingproteinases. In contrast, mumps virus, also utilizing ubiquitous sialic acids as the receptor, causes systemic, multiorgan tropic infection because of its highly cleavable oligobasic F0 motif. Measles virus and others exhibit the same pattern of high cleavability of F0and broad tissue tropism as that of mumps virus. However, respiratory syncytial virus (RSV) and parainfluenzatype 3 (PIV3), whose F0precursors also possess an oligobasic cleavage motif, generally cause localized infection of the respiratory tract. In immunocompromised infants, however, RSV was multiorgan-tropic, and in cyclophosphamidetreated animals, the virus spread outside the respiratory tract and into other organs whereas Sendai virus remained localized (3, 18). It thus appears likely that RSV is potentially pantropic, although immune response localizes the infection. PIV3 has a greater predilection to cause pneumonia than PIV1, which has a single arginine cleavage motif, and even induces a significant level of viremia in humans, an indication of systemic infection (11, 17). The consensus sequence RXK/RR at the cleavage site appears to be the minimumrequirement for recognitionby the ubiquitous proteinases.

(a) Paramyxoviruses The F0of virulent NDV strains is cleaved by a proteinase(s) ubiquitously present throughout the body and the infection is consequently pantropic or systemic, while avirulent strains undergo F0cleavage only in a few limited tissue types expressing a specific protease(s), hence causing an infection localized to particular organs such as the respiratory and alimentary tracts (34, 35, 37). Cleavage of the former type takes

(b) Other VirusFamilies The cleavability of the glycoprotein HA of influenza A virus in orthomyxoviridae determines whether a virus strain has a wide spectrum of target tissues (as virulent avian influenza strains have) or a narrow one (as mammalian and avirulent avian influenza strains have). The cleavage sites of highly cleavable HAs generally consist of multiple basic residues, whereas those of HAs of restricted cleavability consist of single

A

B

Fig.

1.

(A)

Precursor-product

coprotein.

For

teine

residues.

lines

and

those of

age

site.

HAI

and

gp

not

influenza

are

and

relationship see

Disulfide

products

41

details,

the

text.

bridges

defined

determined A HA2

noncovalently

virus are

of

Vertical

by HA

are

dashed and

HIV-1

disulfide-linked,

NDV

bars

fusion

connected lines.

gly-

represent

(B)

gp160. •¥, whereas

cysby

solid

Cleavage cleavgp120

associated.

(Fig. 1A) (16). Many enveloped animal viruses in other diverse taxonomic families were then found to share essentially the same mechanism of cleavage activation with the paramyxovirus family for expressing fusion activity and infectivity. Biologically active cleavage products are illustrated with the two wellstudied examples, influenza A virus hemagglutinin (HA) and the Env protein (gp160) of human immunodeficiencyvirus type 1 (HIV-1) (Fig. 1B).

MINIREVIEW

Table

1. Cleavage

site sequence

and cleavability

arginine residue (Table 1). The consensus RXK/RR cleavage motif is widely found in the highly cleavable group, and its requirement for ubiquitous cleavage has been confirmed by numerous site-directed mutagenesis studies (25). Thus, influenza A virus presents the same pattern of tropism and pathogenicity as described for NDV. One unique feature of an influenza HA (H5) is that a sugar chain attached to a site close to the oligobasic cleavage site sterically interferes with the action of ubiquitous processing enzymes (Table 1, ref. 20). Thus, the point mutation leading to the loss of the glycosylation site greatly increases the viral pathogenicity and was the cause of the 1983 outbreak (20).

3

of viral glycoprotein

precursors

In virtually all cases of the other enveloped viruses, an oligobasic cleavage site exists in the precursor glycoproteins, which are therefore activated in a wide variety of cells in culture and possibly in vivo (32, 33). Among these is the gp160 of HIV-1 in retroviridae (Table 1), whose cleavage to gp120 and gp41 at REKR site is necessary for viral fusion activity and infectivity (27). Although receptors and other host molecules are important for determining HIV-1 tropism, high cleavability of the gp160 is apparently a precondition for the virus to spread efficiently within and between target organs. Various structural features have been revealed by cleavage-site-directed mutagen-

Y. NAGAI

4

esis of HIV-1 gp160 but the RXK/RR motif again seems to be important as the minimum requirement for recognition by the ubiquitously occurring proteinases (4). 4. Virus-Activating Sight

Proteinases

Are Coming into

in

the

middle

phase

Although ovo

the

has

role

not

the

suggesting

tion

the

that

restricted

the

blood-clotting

to

cascade

reac-

side

where

X

(a) Blood-Clotting Factor Xa (FXa) Avirulent NDV, Sendai virus and mammalian and avirulent avian influenza A virus are thought to replicate well in the allantoic and amniotic sacs of the chicken embryo because of the availability of a VAP in the fluids filling these sacs. By using allantoic and amniotic fluids as starting materials, this VAP has been isolated and purified to homogeneity (8, 10). The VAP activates the viruses by cleaving their F0 or HA at a specific single arginine site. The VAP is a calciumdependent serine proteinase consisting of two subunits: a catalytic 33-kDa chain and a 23-kDa chain required for calcium binding. The amino-terminal amino acid sequence of each subunit is highly homologous to that of the activated form (FXa) of human or bovine blood-clotting factor X (FX), a member of the vitamin K-dependent serine proteinases. The identity of the VAP with FXa has been established by comparing the primary structure, deduced from the cloned cDNA sequence encoding the VAP, with those of human and bovine FX (46) and by amino acid sequencing of the FXa isolated from chicken plasma (8). FX is generally synthesized in the liver and circulates as one of the plasma proteinase zymogens. It was therefore an unexpected discovery that not only the liver but also a variety of other chicken embryo tissues, including the allantoic and amniotic cells as well as the kidney, intestine and spleen, express the specific mRNA and the translation product (39). Also surprise is the finding that active FXa is naturally present in the allantoic and amniotic fluids, since it is usually generated only

eny

with

site

also

be

trypsin

activated

or

for

acids

and

factor-like kDa

Ca2+

the

first

domains

in

cleavage

(Table are of

are

susceptible

by

required

than

that

the

of

appears

enzyme

on

the ƒÁ-carboxyglu-

two

amino

It will

action

of

the

1).

FXa

FXa

to

which

cleaved

lower

of

the

the com-

HAs,

sac

bound of

explain

they

concentration

via

often

activation

is much

P3

viral

E/QXR,

different

efficiency

to

bilayer

could

amount

vitro

the

influenza

allantoic

carat

as

virus

some

The

high

the

embryo

This

whether

in

This

attributable

lipid

tamic

the

enzymes.

(8).

be

the

in

learn

activation

trypsin

of

However,

seen in

to

other

virus

(8).

are

with

charge

for

The

replaceable

chicken

of

FXa.

negative

1).

FXa

of

generalized

T (Table

ovo

site

importance

the

poorly in

Interestingly,

site,

of

interesting

FXa

to

S or

sequences

are

in

only

cleavage

be

the

(21).

cleavage

specificity

pared

the

an

FXa

substrate to

than

recognition

is G,

is

activation the

appears

rather

similar

type

which

natural

activated

a

supports

this

chloro-

cleavage

suggesting

glycoproteins have

(E)

is

FXa-

macromolecular

mimics

the

chain

substrate

higher

block CMK

(Q),

bonyl for

can

acid

glutamine

with

further

EGR-CMK),

prothrombin,

glutamic

virus

contact

of

a

observa-

glutamyl-glycyl-arginine

peptidyl

bovine

the Sendai

finding

in

evidence

as

direct

Indeed,

(dansyl

cells,

This

activator

from

well

activation

event.

ketone

as

This

dansylated

penetrates

P3

(39).

fide

established,

in

cleavage

extracellular

a bona

come

NDV

surfaces

fluids

methyl

has

of

tissue

as

fully

this

that

inhibitor,

FXa

been

spread

notion

(8).

of

yet

strongly

containing

Although proteinase-mediated virus activation and its importance for the determination of virus tropism and pathogenicity were already recognized in the mid1970s, it is only recently that the candidate proteinases involved have been identified. A virus-activating proteinase (VAP) catalyzing cleavage at a single arginine site was first isolated from the chicken embryo. Subsequently, a different VAP of the same category has been isolated from the rat lung. The oligobasic RXK/ RR motif is shared by many cellular proproteins (15), and the recent progress in identifying their processing enzymes has greatly helped to characterize this type of VAP.

of

tion.

epidermal

growth

terminus

of

the

23-

chain.

(b)

Tryptase Rats

Tryptase

well

bronchial

nine

and HA,

cleavage

both

to

lung,

resulting

tality

rate

enzyme

(51).

and

its

Thus,

is

in

tryptase for

the

pathogenicity

concentration

required remarkably

of

for high

(at

the

the

for virus

in-

lungs,

lesions

Clara

is

lung.

Sendai

virus

least

1 ƒÊg

this prog-

and

anti-

spreading

lung

rat

and

a single-argi-

in and

the mor-

probably

dissemination in

in

purified Fo

required

virus

a reduction

responsible

not

cells

The

Sendai

lumen blocks

Clara

feature is

virus.

proteinase

virus

activates

the

enzyme in

(24).

rats

which

Clara in

this

of

Calcium

Tryptase

extracellularly

from

Sendai

of

Sendai

serine

secreted

activates

motif.

activation.

serum

and

epithelium

virus

to

arginine-specific in

cleaves

fluenza

vitro

mice

is an

exclusively

enzyme

virus

as

Clara

located the

Clara

as

the

of The

Sendai enzyme

activation ml-1;

ref.

in 24),

MINIREVIEW

when

compared

ref.

8).

with

Such

occur

a high

normally

that

following

are

modulated

viruses

as

to

tion

of

type

tryptase

suppresses

the

Bacterial

of

influenza

cescens

of

can

are

of

or

at

been

related

to

these

mating

di-

proteinimporfactor

physiological influenza

of

Subsequently,

PC2

and

proprotein

fpslfms

these

region

a variety

involved

in

secretory

of

proprotein

pathway. and are

tissues

PC3 localized

is

is

and

cell

processing On

the

other

restricted to

secretory

the and

to

and

organi-

subtilisins in

including

PC1)

a

pro-ƒ¿ RR

homologues

(PC

denotes

a unique

Furin, is

that

cell

yeast

the at

constitute (2).

(fur),

endoproteinase

the

identified, called

subtilisins

upstream

localized

KEX2 been

of

marker

in

prokaryotic

(formerly

convertase);

mammalian

field

sequence

the

been have

genetic

processes

both

have PC3

this

(a

toxin

several

eukaryotes

in

KEX2

pro-killer

with

The

always

proteinase

shares

frequently

involved

which

the

which

therefore

A landmark

cerevisiae, and

are

prohormones.

proteinases

function)

homologies

higher

They

as

motifs

has

the

discovery

killer

and

zational

PC2

motif

the

certain

and

motifs

for.

the

factor sites

in

cleave

Hageman

of

pro-

Endoproteinase

and

Saccharomyces

ed

to

plasma

well

cleavage

proproteins

searched was

of

appear

and

in

The

suggested

as

be mar-

48).

cleavage

bacterial

dibasic

interest

biology

furin,

should

replication (1,

activating results

with

Serratia

virus

activation

cellular

great

(19).

it

bacteria

through

devel-

proteinases

and

single-arginine

by

the infection

context,

cleavage-activation

processing

ly

these

a Subtilisin-Like

in

OKR

and

26).

Furin,

long

infec-

Clara

promote

symptoms

by the

Oligobasic

of

Clara

virus

produce

influenza

Earlier

for

to

this

severer

activation

found

the

(23).

aureus

plasminogen

(1,

Sendai

often

In

indirectly

factors

pul-

endogenous

tryptase

combined

they

promote

zymogens.

HAs

believed

and

with

or

(FXII)

by

surfactant

after

produced

tance

the

progeny that

is an

of

Staphylococcus

HAs

ase

secreted

that

to

seems

conditions of

found

cells,

and

release

producing

teinases

(d)

Clara

it

local

now

is also

categories.

that

rectly

was

ml-1;

appear

but

activation

alveolar

pneumonia

various

mice,

It

production

viruses,

noted

not

infection,

the

bacteria

opment

does

ng

Proteinases

Certain

the

2

30

surface

the

Clara.

stimulates

of

virus

which

as

(about

bronchial

facilitate

tryptase

well

inhibitor

(c)

the

surfactant,

cells

FXa

concentration

on Sendai

by

monary

chicken

fami-

encoded Golgi

by

membrane-

ubiquitously lines in

express-

and the

hand,

is possibly constitutive

expression

neuroendocrine vesicles

the

of cells.

and

consid-

5

ered to be responsible for processing in the regulated secretory pathway. Several lymphoid cell lines including NALM-6 exhibited a reduced cleaving activity even for F0of virulent NDV (33). Most of these cell lines were later found to express little endogenous furin at the transcriptional level (42). F0cleavage in NALM-6 cells is stimulated significantly by coexpressing KEX2 in a recombinant vaccinia virus (33). Similar coexpression studies have further revealed that furin is fully capable of cleaving F0 while PC2 and PC3 are entirely incapable or only partially capable of cleavage, and that the cleavage by furin results in enhancement of viral infectivity (9). Neither furin nor PC2 and PC3 can attack the single-arginine site of avirulent strains (9). Coexpression systems, however, often result in the expression in large excess beyond the basal level, perhaps giving rise to an unphysiological status. On the other hand, normal cells, which are fully capable of cleaving NDV F0 and other substrates at the RXK/RR site, express furin usually at a low level. Thus, the data obtained by coexpression systems should be carefully interpreted. LoVo cells, a cell line from a human colon carcinoma, are unable to process several cellular proproteins with the RXK/RR cleavage-motif, and were found to be defective in furin (29, 47). The normal human fur gene encodes a polypeptide of 794 amino acids which consists of the unique structural and functional domains, including the signal peptide, propeptide, subtilisin-like catalytic domain, homo B domain, cysteinerich domain and transmembrane domain, in this order from the amino terminus. Several independent furin cDNA clones isolated from LoVo cells unexceptionally revealed a one-nucleotide deletion at positions 1,283 to 1,286 (numbered according to ref. 55), where four successive T residues are present in the wild-type cDNA; only three T residues were found in the LoVo cDNAs (47). This deletion of one T residue was deduced to cause a frameshift at amino acid position 479, followed by an aberrant termination in the homo B domain of the furin polypeptide. The requirement of an entire homo B domain for processing activity was already demonstrated by carboxy-terminal truncation analysis (13). LoVo cells were thus expected to complement the previous results obtained by those coexpression studies, and were now found to be unable to cleave and activate the F0precursor of a virulent strain NDV (40). Furthermore, constitutive expression of the intact human furin has rendered LoVo cells fully capable of F0processing (manuscript in preparation). Several influenza HAs with an oligobasic cleavage site behave quite similarly to the NDV F0 in LoVo cells

6

Y. NAGAI

(manuscript in preparation). Thus, these studies using LoVo cells greatly substantiated the role of furin in viral glycoprotein cleavage and activation. A mutant line of Chinese hamster cells which is defective in NDV F0 processing was described, and this genetic lesion was also complemented by expressing mouse furin (28). A biochemical study was attempted to isolate an endoproteinase that activates the influenzafowl plaque virus (FPV) HA from Madin-Darby bovine kidney (MDBK) cells and was able to concentrate the enzyme sufficiently to establish its identity with furin using a specific antiserum (45). It has also been shown that HA cleavage increases in cells where the HA and furin are coexpressed by vaccinia virus vectors. Furthermore, cleavage site-specific decanoyl-REKR-CMK, which is able to penetrate cells, inhibits intracellular cleavage-activation of HA (45). A very similar pattern of increased glycoprotein cleavage by coexpressed furin and/or activation inhibition by decanoyl-REKRCMK has been demonstrated for PIV3 F0 (41) and HIV-1gp160 (12, 30). Recently,Siezen et al (44) presented a model for the three-dimensional structure of the catalytic domain of human furin and its interaction with model substrates. This model predicts distinct pockets for S1, S2 and S4 subsites of the catalytic site, a strong predominance of negatively charged residues in these subsites (Fig. 2), hence their electrostatic interactions with the corresponding positively charged P1, P2 and P4 residues of substrates for specific interaction. Exactly the same charge complementarityis seen between these subsites and corresponding residues of the F0 of virulent NDV and HIV-1 gp160 (Fig. 2). The model further predicts that the S3, 55 and S6 subsites, also negatively charged (Fig. 2), may further contribute to the high substrate specificity of furin (44). In this context, it is noteworthy that the positive P5 and the neutral P3 residues of the F0 are respectively quite fit and at least not disturbing the interactionwith furin, while the negative charge at P3 of gp160 apparently repells the S3 subsite, although the neutral P5 does not appear to be an obstacle (Fig. 2). These features strongly support the highly specific recognition and cleavage of the Fo by furin but may somewhat underestimate the affinity of the gp160 to this enzyme (see below). In addition, HIV-1 gp160 appears to be unique in that there is a cluster of basic residues three residues upstream from the authentic cleavage site (Fig. 2) and that site-directed mutagenesis of these basic residues to neutral residues abolished cleavage-activation (4). The negatively charged P3 of FPV (Table1) is likewise an obstacle in the interaction, but three successive positively charged

Fig. 2. Amino acid residues in subsites constituting the catalytic domain of human furin (44) and the cleavage site sequences of viral substrates. Dashes indicate identity with the above sequence. Negatively charged residues are shown by outlined letters. Positively charged residues in viral sequences are boxed.

residues may facilitate the interaction. Almost complete resistance of avirulent NDV F0 to furin (Fig. 2, also see ref.9) substantiates the importance of basic residues at P2 and perhaps at P5 for recognition by furin. A similar modeling study with trypsin predicted presence of no acidic residues but neutral ones for Si, S2 and S4 subsite (unpublished data). Thus, trypsin has a much broader specificity than furin and is able to cleave the F0 of both virulent and avirulent NDV strains (Fig. 2, also see refs. 34, 35, 40). (e) Is Furin the Only Ubiquitous VAP? As described above, there appears to be increasing evidence supporting the concept that furin is responsible for recognition and cleavage of the RXK/RRmotif. However, the conclusion has mainly come from the observation that furin coexpressed by an appropriate vector promoted cleavage-activation of the respective substrates in normal cells with a basal level of endogenous processing activity. The peptidyl CMKs used are not strictly enzyme-specific but can inactivate any enzyme that recognizes and cleaves the related sequences. Besides, the furin-substrate modeling described above suggested that microheterogeneity around and in the cleavage site sequence including the residue at P3 may affect the affinity to furin. LoVo cells defective in furin and incapable of cleaving the F0 of virulent NDV were recently found to be able to process the gp160 of HIV-1with kinetics comparable to that of normal cells, express the glycoprotein in a biologically active, fusogenic form on their surface and produce fully infectious progeny (40). Another observation, which also does not appear to indicate the requirement of furin for HIV-1 gp160 processing, is that a T-cell line, Molt 4, is fully susceptible to HIV-1, although it expresses little or only a marginal level of furin mRNA and does not efficiently process the F0 of virulent NDV (42). These recent

MINIREVIEW

studies would raise a further need to search for and identify the proteinases involved in HIV-1 gp160 processing rather than supporting the notion that furin is responsible. In this context, the Ca2+-independent26kDa proteinase is of considerable interest because it has been isolated from Molt 4 cells and appears to correctly process the gp160 at least in vitro (22), and because the intracellular gp160 processing appears to occur in low Ca2+milieu where NDV F0processing is inhibited(manuscript in preparation). PACE4 and PC6 are also the members of mammalian subtilisins and their involvement in gp160 processing has been suggested because of their broad tissue distribution, although their expression, just like furin, was found to be very low in Molt 4 cells (42). However, in view of the fact that LoVo furin is defective in the presumptive regulatory domains such as the homo B and the cysteine-rich domains but does retain the intact catalytic domain, the possibility has not been ruled out that gp160 cleavage does not require such regulatory domains at all and therefore occurs in LoVo cells, whereas these domains are absolutely necessary for NDV F0 processing. 5. Outlook Virus spread and tissue tropism depend strongly on the proper match between the cleavability of the viral glycoproteinby an endoproteinase and the availability of the proteinase in the host. The structure of the cleavage site is of prime importance for determining whether the virus is activated in a wide variety of tissues or in particular tissue types. With its ability to recognize the single arginine cleavage site, FXa became the first VAP to be identified, followed by tryptase Clara. The VAPs in this category thus differ depending on the host and/or tissue type. It would be interesting to identify influenza-virus-activatingproteinases in the human respiratory tract. Other plasma proteinases as well as coinfecting bacterial proteinases could be involved. Furthermore, many other proteinases are also recruited to the inflammatoryregion late in infection, which likely act as a VAP,but it would be important to identify the endogenous VAP triggering the initial rounds of virus replication. The ubiquitously occurring VAP that recognizes the oligobasic motif has been identified as furin, but the question remains whether it is the only proteinase of this type. In this context, identification of VAP for HIV-1gp160 is of particular importance. Although our current knowledgeof proteinase-mediated virus activation and its significance for viral pathogenicity is still incomplete, it has already stimulated

7

a variety of new approaches to virus disease control. For instance, the therapeutic potential of specific proteinase inhibitors is being tested, with peptidyl CMKs specific for cleavage site sequences taking the lead (8, 12, 41, 45). Avirulent NDVstrains with a single-arginine motif for proteinaserecognition have already been used routinely for protection of birds against the virulent viruses with a highly cleavable oligobasic motif. This highlights the potential of attenuated live vaccines made by cleavage site-directed mutagenesis. For example, if the oligobasic cleavage sites can be made monobasic, measles virus and mumps virus could be attenuated for use as live vaccines. Human influenza A virus is similar to murine Sendai virus in that both possess a single-arginine motif and target the respiratory tract. In view of the potential as a live vaccine of proteinase-activationmutants of Sendai virus that have a cleavage site susceptible to enzymes other than arginine-specific enzymes (50), it would be worth engineering similar mutants of influenza virus. VAPs undoubtedly are not solely involved in virus activation. So, what is the physiological significance of ectopic FX/FXa expression in the chicken embryo? What physiological roles does tryptase Clara play in the rat lung? Is furin the only processing proteinase in the constitutive secretory pathway in higher eukaryotes? These are new issues raised by the research on VAPsand are well worthy of investigation. I thank uscript eling

K. Kakuta

and A. Mitsuzawa

and Y. Takeuchi of proteinase-substrate

ported by research Science and Culture

for providing

for preparing information

interactions.

grants from the and the Ministry

My

the man-

on the modwork

was

sup-

Ministry of Education, of Health and Welfare,

Japan.

References

1) Akaike, T., Molla, A., Ando, M., Araki, S., and Maeda, H. 1989. Molecular mechanism of complex infection by bacteria and virus analyzed by a model using serratial proteinase and influenza virus in mice. J. Virol. 63: 22522259. 2) Barr, P.J. 1991. Mammalian subtilisins: the long-sought dibasic processing endoproteases. Cell 66: 1-3. 3) Blandford, G. 1975. Studies on the immune response and pathogenesis of Sendai virus infection of mice. III. The effects of cyclophosphamide. Immunology 28: 871-883. 4) Bosch, V., and Pawlita, M. 1990. Mutational analysis of the human immunodeficiency virus type 1 env gene product proteolytic cleavage site. J. Virol. 64: 2337-2344. 5) Buckland, R., and Wild, F. 1989. Leucine zipper motif extends. Nature 338: 547. 6) Chambers, P., Pringle, C.R., and Easton, A.J. 1990. Heptad repeat sequences are located adjacent to hydropho-

8

Y. NAGAI

bic

regions

Gen. 7)

in

Virol.

Gething,

the

White,

of

the

B.,

J.M.,

nant

Natl.

M.,

homologous

and

glycoprotein.

Waterfield, of

J.

Sci.

T.,

blood

viral

tropism

in

B.,

Ohnishi,

T.,

Y.

chick

precursor

22)

2737-2740.

Inocencio,

1990.

clotting

analysis

during 75:

An

factor

N.M.,

endoprotease

X

embryo.

21)

1978.

virus:

U.S.A.

Toyoda,

Nagai,

the

M.D.

Sendai

generated

Acad.

and

to

of

fusion

protein

Ogasawara,

Hamaguchi,

virus

sequence

Proc.

Gotoh,

of

fusion

NH2-terminal

activation. 8)

types

3075-3080.

M.J.,

Purification of

several

71:

as

a

EMBO

determi-

J.

9:

4189-

23)

4195. 9)

Gotoh, Nakayama,

K.,

Mammalian vating ity

Barr,

P.J.,

Inocencio,

the

to

G.,

and

fusion PC2

or

Esaki,

Nagai,

proteinases

paramyxovirus

furin/PACE

N.M.,

Thomas,

subtilisin-related of

of

Y.,

in

Y.

J.

1992.

cleavage

glycoprotein:

PC1/PC3.

E.,

acti-

24)

superior-

Virol.

66:

6391-

6397. 10)

Gotoh,

B.,

1992.

Isolation

amniotic

11)

FEBS

Gross,

P.A.,

Rev.

H.-D.,

and

cleavage

13)

with

W.

1992.

of

HIV-1

M.,

growth

of

tural

and

culture

cells.

Hosaka,

M.,

Nakayama, for

K.

and

man.

160

by

spe-

K.

furin,

27)

358-361.

a

cleavage

tissue

Klenk,

furin-mediated

360:

1992.

Kex2 at

like

Arg-X-

28)

296-301. Trypsin

action

culture

cells.

grown

in

on

III.

eggs

the

Struc-

and

tissue

1457-1465. M.,

Kim,

W.-S.,

J.,

Watanabe,

Murakami,

Arg-X-Lys/Arg-Arg by

pathway.

J.

furin

Biol.

T.,

K., motif

catalyzed

secretory

E.,

gp

of

viruses

cleavage

Per-

in

Nakayama,

1973.

Ikemizu,

1991.

precursor

stitutive

in

12:

K.,

of

Nature

111:

M.

Sendai

Virol.

virus

Shaw,

Inhibition

precursor

Nagahama,

Hatsuzawa,

H.,

properties

virus

J.

3

glycoprotein

J. Biochem.

of

1973.

26)

Angliker,

in

Ohuchi,

Sendai

difference

acti-

M.G.M. type

K.,

enzymatic

sites.

Homma,

the

25) Curnen,

ketones.

involved

Lys/Arrg-Arg

15)

V.,

Murakami,

and

Y.

as

894-898.

Garten,

endoprotease

Nagai,

embryo

paramyxovirus

parainfluenza 108:

Bosch,

K.,

for

and

peptidyl-chloromethyl

Molecular

14)

R.H.,

activation

and

chick

274-278.

S.,

Hatsuzawa,

T.,

from

responsible

Dis.

Hallenberger,

Xa

296:

Green,

Resp.

Ogasawara,

factor

Lett.

infection

Am.

cific

F.,

of

endoprotease

vation.

sistent

12)

Yamauchi,

as

within

Chem.

a signal the

266:

con-

12127-

30)

12130. 16)

Iwata,

S.,

Gotoh,

Schmidt,

B.,

ment

of

Sendai 17)

Johnson,

J. Virol.

R.A.,

Immun. Julius, ner, the

J.

Kawaoka, virulence

Nagai,

37:

369-373. Brake,

1984.

Isolation

R.H.

of

1973. of

Suffin,

1982.

S.C.,

Respiratory

Y., of

Blair, of

the

cleaving yeast Naeve, H5N2

1994.

H.,

Assignof

Viremia

during

hamsters.

Proc.

31)

Horswood,

R.L.,

32)

745-748.

G.A.,

A.,

Kido,

glycoprotein

syncytial

cyclophosphamide-treated

D.A.,

M., Y.

fusion

infection

144:

R.M.

in

lysine-arginine

processing 20)

Med.

Suzuki,

3200-3206.

virus

Prince,

Chanock,

infection

19)

Biol.

the

Green, 3

K.,

and in

68:

and type

Exp.

and

M., bridges

D.P.,

Johnson,

Titani,

Hamaguchi,

virus.

Soc.

A.C.,

disulfide

parainfluenza

18)

29)

and

cotton

L.,

C.W.,

R.,

structural

endopeptidase

prepro-ƒ¿-factor.

influenza

Kunisawa,

putative

Cell and viruses

Webster, in

virus

rats.

Infect.

and

Thor-

gene required

37:

for for

1075-1089. R.G.

chickens

1984. associat-

Is

ed with loss of carbohydrate from hemagglutinin? Virology 139: 306-316. Kettner, C., and Shaw, E. 1981. The selective affinity labeling of factor X by peptides of arginine chloromethyl ketone. Thromb. Res. 22: 645-652. Kido, H., Kamoshita, K., Fukutomi, A., and Katunuma, N. 1993. Processing protease for gp160 human immunodeficiency virus type I envelope glycoprotein precursor in human T4 + lymphocytes. J. Biol. Chem. 268: 1340613413. Kido, H., Sakai, K., Kishino, Y., and Tashiro, M. 1993. Pulmonary surfactant is a potential endogenous inhibitor of proteolytic activation of Sendai virus and influenza A virus FEBS Lett. 32: 115-119. Kido, H., Yokogoshi, Y., Sakai, K., Tashiro, M., Kishino, Y., Fukutomi, A., and Katunuma, N. 1992. Isolation and characterization of a novel trypsin-like protease found in rat bronchiolar epithelial Clara cells: a possible activator of the viral fusion glycoprotein. J. Biol. Chem. 267: 13573-13579. Klenk, H.-D., and Garten, W. 1994. Host cell proteases controlling virus pathogenicity. Trends Microbiol. 2: 3943. Lazarowitz, S.G., Goldberg, A.R., and Choppin, P.W. 1973. Proteolytic cleavage by plasmin of the HA polypeptide of influenza virus: host cell activation of serum plasminogen. Virology 56: 172-180. McCune, J.M., Rabin, L.B., Feinberg, M.B., Lieberman, M., Kosek, J.C., Reyes, G.R., and Weissman, I.L. 1988. Endoproteolytic cleavage of gp160 is required for the activation of human immunodeficiency virus. Cell 53: 55-67. Moehring, J.M., Inocencio, N.M., Robertson, B.J., and Moehring, T.J. 1993. Expression of mouse furin in a Chinese hamster cell resistant to pseudomonas exotoxin A and viruses complements the genetic lesion. J. Biol. Chem. 288: 2590-2594. Mondino, A., Giordano, S., and Comoglio, P.M. 1991. Defective posttranslational processing activates the tyrosine kinase encoded by the MET proto-oncogene (hepatocyte growth factor receptor). Mol. Cell. Biol. 11: 60846092. Morikawa, Y., Barsov, E., and Jones, I. 1993. Legitimate and illegitimate cleavage of human immunodeficiency virus glycoproteins by furin. J. Virol. 67: 3601-3604. Morrison, T.G., Ward, L.J., and Semerjian, A. 1989. Intracellular processing of the Newcastle disease virus fusion glycoprotein. J. Virol. 53: 851-857. Nagai, Y. 1993. Protease-dependent virus tropism and

pathogenicity. Trends Microbiol. 1: 81-87. 33) Nagai, Y., Inocencio, N.M., and Gotoh, B. 1991. Paramyxovirus tropism dependent on host proteases activating the viral fusion glycoprotein. Behring Inst. Mit. 89: 3545. 34) Nagai, Y., and Klenk, H.-D. 1977. Activation of precursors to both glycoproteins of Newcastle disease virus by proteolytic cleavage. Virology 77: 125-134. 35) Nagai, Y., Klenk, H.-D., and Rott, R. 1976. Proteolytic cleavage of the viral glycoproteins and its significance for the virulence of Newcastle disease virus. Virology 72:

MINIREVIEW

494-508. 36) Nagai, Y., Ogura, H., and Klenk, H.-D. 1976. Studies on the assembly of the envelope of Newcastle disease virus. Virology 69: 523-538. 37) Nagai, Y., Shimokata, K., Yoshida, T., Hamaguchi, M., Iinuma, M., Maeno, K., Matsumoto, T., Klenk, H.-D., and Rott, R. 1979. The spread of a pathogenic and an apathogenic strain of Newcastle disease virus in the chick embryo as depending on the protease sensitivity of the virus glycoproteins. J. Gen. Virol. 45: 263-272. 38) Neyt, C., Geliebter, J., Slaoui, M., Morales, D., Meulemans, G., and Burny, A. 1989. Mutations located on both Fl and F2 subunits of the Newcastle disease virus fusion

39)

40)

41)

42)

43)

44)

protein confer resistance to neutralization with monoclonal antibodies. J. Virol. 63: 952-954. Ogasawara, T., Gotoh, B., Suzuki, H., Asaka, J., Shimokata, K., Rott, R., and Nagai, Y. 1992. Expression of factor X and its significance for the determination of paramyxovirus tropism in the chick embryo. EMBO J. 11: 467-472. Ohnishi, Y., Shioda, T., Nakayama, K., Iwata, S., Gotoh, B., Hamaguchi, M., and Nagai, Y. 1994. A furin-defective cell line is able to process correctly the gp160 of human immunodeficiency virus type 1. J. Virol. 68: 4075-4079. Ortmann, D., Ohuchi, M., Angliker, H., Shaw, E., Garten, W., and Klenk, H.-D. 1994. Proteolytic cleavage of wild type and mutants of the F protein of human parainfluenza virus type 3 by two subtilisin-like endoproteases, furin and Kex2. J. Virol. 68: 2772-2776. Sakaguchi, T., Fujii, Y., Kiyotani, K., and Yoshida, T. 1994. Correlation of proteolytic cleavage of paramyxovirus F protein precursors with expression of the fur, PACE4 and PC6 genes in mammalian cells. J. Gen. Virol. in press. Scheid, A., and Choppin, P.W. 1974. Identification of biological activities of paramyxovirus glycoproteins. Activation of cell fusion, hemolysis, and infectivity by proteolytic cleavage of an inactive precursor protein of Sendai virus. Virology 57: 475-490. Siezen, R.J., Creemers, J.W.M., and Van De Ven, W.J.M. 1994. Homology modelling of the catalytic domain of human furin. A model for the eukaryotic subtilisin-like

proprotein convertases. Eur. J. Biochem. 222: 255-266. 45) Steineke-Grober, A., Vey, M., Angliker, H., Shaw, E., Thomas, G., Roberts, C., Klenk, H.-D., and Garten, W. 1992. Influenza virus hemagglutinin with multibasic

9

cleavage site is activated by furin, a subtilisin-like endoprotease. EMBO J. 11: 2407-2414. 46) Suzuki, H., Harada, A., Hayashi, Y., Wada, K., Asaka, J., Gotoh, B., Ogasawara, T., and Nagai, Y. 1991. Primary structure of the virus activating protease from chick embryo. Its identity with the blood clotting factor Xa. FEBS Lett. 283: 281-285. 47) Takahashi, S., Kasai, K., Hatsuzawa, K., Kitamura, N., Misumi, Y., Ikehara, Y., Murakami, K., and Nakayama, K. 1993. A mutation of furin causes the lack of precursor-

48)

49)

50)

51)

processing activity in human colon carcinoma LoVo cells. Biochem. Biophys. Res. Commun. 195: 1019-1026. Tashiro, M., Ciborowski, P., Klenk, H.-D., Pulverer, G., and Rott, R. 1987. Role of staphylococcus protease in the development of influenza pneumonia. Nature 325: 536537. Tashiro, M., and Homma, M. 1983. Pneumotropism of Sendai virus in relation to protease mediated activation in mouse lung. Infect. Immun. 39: 879-888. Tashiro, M., and Homma, M. 1985. Protection of mice from wild-type Sendai virus infection by a trypsin-resistant mutant, TR-2. J. Virol. 53: 228-234. Tashiro, M., Yokogoshi, Y., Tobita, K., Seto, J.T., Rott, R., and Kido, H. 1992. Tryptase Clara, an activating protease for Sendai virus in rat lungs, is involved in pneumopatho-

genicity. J. Virol. 66: 7211-7216. 52) Toyoda, T., Gotoh, B., Sakaguchi, T., Kida, H., and Nagai, Y. 1988. Identification of amino acids relevant to three antigenic determinants on the fusion protein of Newcastle disease virus that are involved in fusion inhibition and neutralization. J. Virol. 62: 4427-4430. 53) Toyoda, T., Sakaguchi, T., Hirota, H., Gotoh, B., Kuma, K., Miyata, T., and Nagai, Y. 1989. Newcastle disease virus evolution. II. Lack of gene recombination in generating virulent and avirulent strains. Virology 169: 273-282. 54) Toyoda, T., Sakaguchi, T., Imai, K., Inocencio, N.M., Gotoh, B., Hamaguchi, M., and Nagai, Y. 1987. Structural comparison of the cleavage-activation site of the fusion glycoprotein between virulent and avirulent strains of Newcastle disease virus. Virology 158: 242-247. 55) van den Ouweland, A.M.W., van Duijnhoven, H.L.P., Keizer, G.D., Dorssers, L.C.J., and Van de Ven, W.J.M. 1990. Structural homology between the human fur gene product and the subtilisin-like protease encoded by yeast KEX2. Nucleic Acids Res. 18: 664.