ArticlePDF Available

Tracking familial transmission of Kaposi's sarcoma-associated herpesvirus using restriction fragment length polymorphism analysis of latent nuclear antigen

Authors:
  • Kamuzu University of Health Sceinces (KUHeS)

Abstract and Figures

Intra-familial transmission of Kaposi's sarcoma associated herpesvirus (KSHV) is likely to occur in geographical regions where KSHV infection is highly endemic. Transmission has been studied previously indirectly using serological techniques, however direct documentation of specific transmission routes has yet to be reported. The internal repeat domain (IRD) of the KSHV opening reading frame (ORF) 73 was shown previously to exhibit restriction-fragment length polymorphism (RFLP). Analysis of such polymorphism was undertaken using nested ORF 73 IRD PCR products derived from the blood and mouth rinse samples of individuals in Malawian family groups. The resulting RFLP patterns were unique to an individual and could be compared between family members. In three of eight families studied, identical RFLP patterns were recovered from family members; in the remaining five families, dissimilar RFLP patterns were revealed. Results from RFLP analysis were compared to sequencing data recovered from family members for the first variable region of the hypervariable KSHV ORF K1. Patterns of intra- and extra-familial transmission inferred from ORF K1 sequencing data were corroborated mainly using ORF 73 IRD RFLP analysis.
Content may be subject to copyright.
Journal of Virological Methods 105 (2002) 297303
Tracking familial transmission of Kaposi’s
sarcoma-associated herpesvirus using restriction fragment
length polymorphism analysis of latent nuclear antigen
R.D. Cook
a,b,
*, T.A. Hodgson
a
, E.M. Molyneux
c
, E. Borgstein
d
,
S.R. Porter
a
, C.G. Teo
b
a
Department of Oral Medicine,Eastman Dental Institute for Oral Health Care Sciences,Uni6ersity College London,London,UK
b
Central Public Health Laboratory,Virus Reference Di6ision,
61
Colindale A6enue,London NW
95
HT,UK
c
Department of Paediatrics,College of Medicine,Uni6ersity of Blantyre,Blantyre,Malawi
d
Department of Surgery,College of Medicine,Uni6ersity of Blantyre,Blantyre,Malawi
Received 13 February 2002; received in revised form 27 May 2002; accepted 28 May 2002
Abstract
Intra-familial transmission of Kaposi’s sarcoma associated herpesvirus (KSHV) is likely to occur in geographical
regions where KSHV infection is highly endemic. Transmission has been studied previously indirectly using
serological techniques, however direct documentation of specific transmission routes has yet to be reported. The
internal repeat domain (IRD) of the KSHV opening reading frame (ORF) 73 was shown previously to exhibit
restriction-fragment length polymorphism (RFLP). Analysis of such polymorphism was undertaken using nested
ORF 73 IRD PCR products derived from the blood and mouth rinse samples of individuals in Malawian family
groups. The resulting RFLP patterns were unique to an individual and could be compared between family members.
In three of eight families studied, identical RFLP patterns were recovered from family members; in the remaining five
families, dissimilar RFLP patterns were revealed. Results from RFLP analysis were compared to sequencing data
recovered from family members for the first variable region of the hypervariable KSHV ORF K1. Patterns of intra-
and extra-familial transmission inferred from ORF K1 sequencing data were corroborated mainly using ORF 73 IRD
RFLP analysis. © 2002 Elsevier Science B.V. All rights reserved.
Keywords
:
Kaposi’s sarcoma associated herpesvirus; Restriction fragment length polymorphism; Familial transmission; Oral
www.elsevier.com/locate/jviromet
1. Introduction
Kaposi’s sarcoma associated herpesvirus
(KSHV) is linked causally to all epidemiological
forms of Kaposi’s sarcoma (KS) (Boshoff and
Weiss, 2001). The prevalence of KSHV infection,
as measured by serology, is low in North America
* Corresponding author. Tel.: +44-208-200-4400x3234; fax:
+44-208-200-1569
E-mail address
:
rck@phls.org.uk (R.D. Cook).
0166-0934/02/$ - see front matter © 2002 Elsevier Science B.V. All rights reserved.
PII: S0166-0934(02)00123-4
R.D.Cook et al.
/
Journal of Virological Methods
105 (2002) 297303
298
and Europe, where 05% of blood donor groups
are KSHV-antibody positive (Gao et al., 1996;
Simpson et al., 1996). However, in areas of sub-
Saharan Africa, KSHV sero-prevalence levels of
50% or greater have been reported previously
(Gao et al., 1996; Sitas et al., 1999; Rezza et al.,
2000).
Sexual transmission of KSHV is implicated
among homosexual men in North America and
Europe (Martin et al., 1998; Blackbourn et al.,
1999; Dukers et al., 2000), in whom the incidence
of Kaposis sarcoma and the prevalence of KSHV
infection is higher (Beral et al., 1990; Gao et al.,
1996; Kedes et al., 1996; Simpson et al., 1996)
than the general population. In Africa, where
KSHV infection in children is common and where
KSHV sero-prevalence increases with age (Olsen
et al., 1998; Gessain et al., 1999; Rezza et al.,
2000) non-sexual transmission of KSHV within
the family is suspected to occur (Bourboulia et al.,
1998; Gessain et al., 1999; Sitas et al., 1999;
Plancoulaine et al., 2000). The vehicle for such
non-sexual transmission is likely to be oral uids
as KSHV DNA has been detected in both the oral
mucosa (Pauk et al., 2000) and saliva (Koelle et
al., 1997).
Despite multiple serological studies, direct evi-
dence of KSHV transmission has not been
demonstrated. KSHV DNA sequence analysis can
determine viral subtype (Zong et al., 2002), but
the methods involved are time consuming. Re-
cently, a rapid restriction fragment length poly-
morphism analysis of KSHV derived PCR
products (PCR-RFLP) was reported (Zhang et
al., 2000). PCR-RFLP targets the internal repeat
domain (IRD) of KSHV opening reading frame
(ORF) 73, a region of signicant polymorphism
which encodes the latent nuclear antigen (Rain-
bow et al., 1997). Numerous point mutations and
deletions concentrated mainly in the second of
three repeat domains in the ORF 73 IRD result in
size and sequence polymorphism. The objective of
this study was to analyse samples from patients
with Kaposis sarcoma and their family members
from Malawi to determine whether PCR-RFLP
subtyping could be applied to study familial trans-
mission of KSHV.
2. Materials and methods
2
.
1
.Study group
Twenty-two patients attending the Central Hos-
pital, Blantyre, Malawi with cutaneous or oral
mucosal Kaposis sarcoma screened to be sero-
positive for KSHV by an immunouorescence
assay (IFA) (Advanced Biotechnologies Incorpo-
rated, MD) were selected as the index cases. A
further 67 family members of the index cases
without Kaposis sarcoma were invited to join the
study. Following informed consent from each
member of the study group, venous blood and
mouth rinses were obtained.
2
.
2
.DNA extraction
DNA was extracted from the immunomagneti-
cally selected CD45+leukocyte fraction of blood
samples as described previously (Leao et al.,
2000). DNA was extracted from the mouth rinse
samples using a guanidium thiocyanate-silica pro-
cedure (Boom et al., 1990).
2
.
3
.KSHV IRD PCR
PCR amplication of the IRD region of ORF
73 was carried out as described previously (Zhang
et al., 2000) with one minor variation. In each
single round PCR reaction, 2 U of Platinum Taq
DNA polymerase (Invitrogen, Paisley, UK) was
used instead of 1 U to ensure a consistently
positive PCR result.
Using the single round PCR described above,
none or too little PCR product was occasionally
generated from our samples. To increase the sen-
sitivity of PCR detection, a nested PCR was
developed, using as outer primers IRD1-F 5%
ACGCCAACCGCCTACATCT 3%and IRD1-R
5%TCATGTGTGCTAACAACAGG 3%and the
original single round primers described by Zhang
et al. (2000) as second round primers. Both
rounds of PCR were carried out in a 25 ml reac-
tion mixture containing 2 ml extracted DNA, 1.25
U Platinum Taq DNA polymerase, 100 mM each
dNTP, 50 pM each primer, 1.5 mM MgCl
2
,50
mM KCl, 20 mM TrisHCl (pH 8.4) and 2 ×
R.D.Cook et al.
/
Journal of Virological Methods
105 (2002) 297303
299
PCRx enhancer solution (Invitrogen, UK). Ther-
mocycling conditions were as previously described
(Zhang et al., 2000). All samples negative after
single round PCR were repeated using the nested
PCR procedure.
2
.
4
.Nested IRD PCR optimisation
Initially nested PCR using the same conditions
as described for single round PCR resulted in high
molecular weight smearing instead of the expected
nested product. Various strategies to clarify the
second round product included reduced primer
concentration, 25 vs. 35 rounds of thermocycling
in the second round of PCR, titration of key
reagents and reduction in the amount of Taq
polymerase used. A clear second round product
could be visualised after titration of the PCRx
enhancer solution supplied with Platinum Taq
polymerase. Using this reagent at a 2×concentra-
tion in both rounds of PCR yielded a specic
product of the correct size when applied to a
KSHV infected BC-1 cell line extract.
When nested PCR was applied to patient sam-
ples, a non-specic or smeared second round
product was occasionally observed. Dilution of
the rst round product 10- to 20-fold resulted in a
specic second round product in these cases. In
some cases, reducing the number of thermocycling
rounds to 25 improved second round PCR results.
2
.
5
.RFLP
For the RFLP, PCR products generated by
single or nested PCR were digested for 90 min at
37 °C with BanII and MboI restriction enzymes
followed by 30 min at 85 °C to inactivate the
enzymes. RFLP products were visualised with
ethidium bromide using UV transillumination af-
ter electrophoresis through a 1.5% agarose gel.
3. Results and discussion
Based on sequencing data from KSHV infected
BC-1 and PK-1 cell lines, Zhang et al. (2000)
characterised four possible RFLP subtypes; dele-
tions in the IRD resulting in the loss of expected
restriction sites created the different RFLP pat-
terns. Thus, in the KSHV IRD of the BC-1 cell
line, there are ve possible BanII restriction sites
resulting in three bands that can be visualised and
one MboI restriction site resulting in two visible
bands. Subtype 1 samples have an IRD with both
BanII and MboI restriction sites, resulting in
bands of 655, 476/457 and 192 bp in length.
Subtype 2 samples have at least two BanII sites
but no MboI site, resulting in three visible bands
at :1121, 457 and 192 bp. There is only one
BanII site in subtype 3 samples, resulting in two
bands at 1131 and 457 bp, while subtype 4 has
one of each restriction site, resulting in three
visible bands with the 192-bp band missing.
Previously, no correlation has been demon-
strated between these four RFLP subtypes and
the previously described KSHV subtypes (AC)
based on sequencing of the KS330 fragment of
ORF 26 (Zhang et al., 2000). ORF K1 sequencing
revealed that the Malawian KSHV samples be-
long to either the A5 or B1 subtype (Cook et al.,
2002). No correlation was found between these
two ORF K1 subtypes and the RFLP subtypes 2
and 3 recovered in this sample group (Table 1).
Zhang et al. (2000) found that RFLP patterns
from multifocal Kaposis sarcoma lesions in a
single patient were invariant. This study of
Malawian individuals identied one patient with
Kaposis sarcoma, K
i
, from whom KSHV DNA
could be amplied in both blood and mouth rinse
samples. In this patient, the two samples revealed
an identical RFLP pattern, indicating that the
same KSHV variant is carried in both the blood
and the oral compartments. However, samples
from only one subject were available for this
analysis and the ndings should be interpreted
with caution.
It was possible to compare RFLP patterns be-
tween two or more family members in eight of the
family groups. Five additional samples with no
matching family members were also included in
the RFLP analysis to determine the overall distri-
bution of subtypes present in this population. The
results of the single round or nested PCR to
amplify the IRD region of ORF 73 are shown in
Fig. 1. The previously described KSHV nuclear
antigen typing (KVNA typing) procedure (Gao et
R.D.Cook et al.
/
Journal of Virological Methods
105 (2002) 297303
300
al., 1999) could be applied directly to these sam-
ples and we detected size polymorphism in the
IRD between samples in families B, G and W.
However, the IRD products for all other families
appeared identical.
RFLP analysis of all samples is represented in
Fig. 2. Distinct RFLP patterns could be differen-
tiated in the samples from Malawian individuals,
allowing us to make comparisons between family
members. In four families (A, E, H, K) identical
RFLP patterns were present suggesting familial
transmission of KSHV had occurred in these fam-
ily groups. In families A and K, identical RFLP
results were found between a mother (A1/K
i
)and
her children, a son (A2/K
1
) and daughter (A
i
).
Identity could also be found between siblings;
brothers E6 and H2 carried the same RFLP pat-
terns as their corresponding sisters, E2 and H1.
In families B, G, E, T and W, non-identical
RFLP patterns were discovered. In two of these
families (T and G), subtypes 2 and 3 existed in
each family, while in families B, E and W, all
samples were of the same subtype but with differ-
ing RFLP band sizes. Studied in family B were
Table 1
Characteristics of the patients with Kaposis sarcoma and family members involved in the study
Kaposis sarcomaIndividual Relationship PCR-RFLPAge/sex ORF K1
(Yes/No) subtypegenotype
2Yes Daughter 7y/FBA
i
(blood)
2No Mother 34y/FN/A*A1
2N/A*No 10y/MA2 Son
B1 2NoB3 Brother 12y/M
No BrotherB5 7y/MB1 2
No SisterE4 6y/FB1 3
3No Brother 13y/MB1E5
No BrotherE6 10y/MB1 3
E2 No Sister 9y/FN/A* 3
G1 2A532y/MFatherNo
323y/MA5SonNoG2
G
i
(blood) 3Yes Mother 34y/FN/A*
No SisterH1 13y/F3N/A*
N/A* 3BrotherH2 12y/MNo
A5K
i
,K
i
(blood) 30y/FMother 2Yes
SonNoK1 12y/MA5 2
T2 3No Brother 11y/MB1
No Brother 9y/MT3 B1 2
3B120y/MW2 BrotherNo
No Sister 13y/FW4 B1 3
C
i
Yes Son 8y/MB1 3
D1 No Daughter 18/FA5 2
2N/A*30y/MP
i
FatherYes
No 3Father 44y/MX1 B1
No A5Z2 22y/FMother 2
All samples are derived from mouth rinses unless otherwise noted. ORF K1 genotype assignments were determined by sequence
alignment with sequences of known subtype deposited in GenBank and by the presence of characteristic amino acid motifs in the
VR1 and VR2 region of ORF K1 (Zong et al., 1999, 2002). PCR-RFLP subtype assignment was as determined by Zhang et al.
(2000).
* Denotes ORF K1 DNA could not be amplied from these samples (Cook et al., 2002).
R.D.Cook et al.
/
Journal of Virological Methods
105 (2002) 297303
301
Fig. 1. ORF 73 IRD PCR results from (A) samples A1H2
and (B) samples K
i
P
i
. Lanes marked BL correspond to blank
lanes. Samples were amplied using either single round or
nested PCR and were visualised on a 1% agarose gel alongside
a 1 Kb DNA marker.
KSHV DNA could not be amplied (Cook et al.,
2002) it cannot be determined whether or not they
were infected by other family members. However,
a comparison could be made between previously
obtained sequencing data (Cook et al., 2002) from
the rst variable region of the hypervariable ORF
K1 and RFLP patterns for six of the eight
families in this study (B, E, G, K, T, W). RFLP
analysis corroborated with ORF K1 sequence
identity found in families E and K and non-iden-
tity found in families B, E, T and W. Previous
evidence of ORF K1 sequence identity was not
conrmed in family G by RFLP (Fig. 2).
In all families except family G and W (W2), the
children were 13 years of age or younger, consis-
tent with KSHV acquisition through a non-sexual
route. In Africa, where the predominant mode of
KSHV transmission is non-sexual, saliva is likely
to be an important vehicle. Saliva has been de-
tected at high titre in American homosexual men
with and without Kaposis sarcoma (Koelle et al.,
1997) and in mouth rinses from Zimbabwean
women with Kaposis sarcoma (Lampinen et al.,
2000). Of the samples included in this study, 23 of
26 were mouth rinses and all but three of these
were derived from family members without Kapo-
sis sarcoma. The PCR-RFLP assay was able to
reveal size and sequence polymorphism in these
mouth rinse samples from individuals without
Kaposis sarcoma. Subtyping through such a
PCR-RFLP approach should enable KSHV trans-
mission in other endemic populations to be simi-
larly studied.
Acknowledgements
We thank S.-J. Gao of the University of Texas,
Health Science Center for helpful comments con-
cerning this study and A.C.W. Waugh for assis-
tance in the collection and processing of clinical
samples. Ethical approval was obtained prior to
commencing this study from Eastman Dental In-
stitute (UK), University College London (UK)
and the University of Malawi. This work was
supported by Grant DE12176-03 from the Na-
tional Institute of Health.
brothers (B3 and B5), in family T, brothers (T2
and T3), in family E, a sister (E4) and her broth-
ers (E2, E5 and E6), and in family G, a father
(G1), a mother (G) and their son (G2). In family
E, RFLP patterns from siblings E4 and E5 dif-
fered not only from each other, but also from
those of their siblings E2 and E6.
The dissimilar RFLP patterns recovered from
each of these family members indicates that they
may not have acquired KSHV from each other.
Nevertheless, as the subjects studied here were
part of a larger immediate family from whom
Fig. 2. PCR-RFLP analysis of the ORF 73 IRD PCR prod-
ucts from (A) samples A1H2 and (B) samples K
i
P
i
. PCR-
RFLP subtype assignments are indicated beneath each lane.
Lanes marked BL correspond to blank lanes. Samples were
digested using MboIandBanII restriction enzymes and visu-
alised on a 1.5% agarose gel alongside a 100 bp ladder DNA
marker to estimate fragment size.
R.D.Cook et al.
/
Journal of Virological Methods
105 (2002) 297303
302
References
Beral, V., Peterman, T.A., Berkelman, R.L., Jaffe, H.W., 1990.
Kaposis sarcoma among persons with AIDS: a sexually
transmitted infection? Lancet 335, 123128.
Blackbourn, D.J., Osmond, D., Levy, J.A., Lennette, E.T.,
1999. Increased human herpesvirus 8 seroprevalence in
young homosexual men who have multiple sex contacts
with different partners. J. Infect. Dis. 179, 237239.
Boom, R., Sol, C.J., Salimans, M.M., Jansen, C.L., Wertheim-
van Dillen, P.M., van der, N.J., 1990. Rapid and simple
method for purication of nucleic acids. J. Clin. Microbiol.
28, 495503.
Boshoff, C., Weiss, R.A., 2001. Epidemiology and pathogene-
sis of Kaposis sarcoma-associated herpesvirus. Phil.
Trans. R. Soc. Lond. B. Biol. Sci. 356, 517534.
Bourboulia, D., Whitby, D., Boshoff, C., Newton, R., Beral,
V., Carrara, H., Lane, A., Sitas, F., 1998. Serologic evi-
dence for mother-to-child transmission of Kaposi sarcoma-
associated herpesvirus infection. J. Am. Med. Assoc. 280,
3132.
Cook, R.D., Hodgson, T.A., Waugh, A.C.W., Molyneux,
E.M., Borgstein, E., Sherry, A., Teo, C.G., Porter, S.R.,
2002. Mixed patterns of transmission of human her-
pesvirus-8 (Kaposis sarcoma associated herpesvirus) in
Malawian families. J. Gen. Virol. 83, 16131619.
Dukers, N.H., Renwick, N., Prins, M., Geskus, R.B., Schulz,
T.F., Weverling, G.J., Coutinho, R.A., Goudsmit, J., 2000.
Risk factors for human herpesvirus 8 seropositivity and
seroconversion in a cohort of homosexual men. Am. J.
Epidemiol. 151, 213224.
Gao, S.J., Kingsley, L., Li, M., Zheng, W., Parravicini, C.,
Ziegler, J., Newton, R., Rinaldo, C.R., Saah, A., Phair, J.,
Detels, R., Chang, Y., Moore, P.S., 1996. KSHV antibod-
ies among Americans, Italians and Ugandans with and
without Kaposis sarcoma. Nat. Med. 2, 925928.
Gao, S.J., Zhang, Y.J., Deng, J.H., Rabkin, C.S., Flore, O.,
Jenson, H.B., 1999. Molecular polymorphism of Kaposis
sarcoma-associated herpesvirus (Human herpesvirus 8) la-
tent nuclear antigen: evidence for a large repertoire of viral
genotypes and dual infection with different viral genotypes.
J. Infect. Dis. 180, 14661476.
Gessain, A., Mauclere, P., van Beveren, M., Plancoulaine, S.,
Ayouba, A., Essame-Oyono, J.L., Martin, P.M., de The,
G., 1999. Human herpesvirus 8 primary infection occurs
during childhood in Cameroon, Central Africa. Int. J.
Cancer 81, 189192.
Kedes, D.H., Operskalski, E., Busch, M., Kohn, R., Flood, J.,
Ganem, D., 1996. The seroepidemiology of human her-
pesvirus 8 (Kaposis sarcoma-associated herpesvirus): dis-
tribution of infection in KS risk groups and evidence for
sexual transmission. Nat. Med. 2, 918924.
Koelle, D.M., Huang, M.L., Chandran, B., Vieira, J., Piep-
korn, M., Corey, L., 1997. Frequent detection of Kaposis
sarcoma-associated herpesvirus (human herpesvirus 8)
DNA in saliva of human immunodeciency virus-infected
men: clinical and immunologic correlates. J. Infect. Dis.
176, 94102.
Lampinen, T.M., Kulasingam, S., Min, J., Borok, M.,
Gwanzura, L., Lamb, J., Mahomed, K., Woelk, G.B.,
Strand, K.B., Bosch, M.L., Edelman, D.C., Constantine,
N.T., Katzenstein, D., Williams, M.A., 2000. Detection of
Kaposis sarcoma-associated herpesvirus in oral and geni-
tal secretions of Zimbabwean women. J. Infect. Dis. 181,
17851790.
Leao, J.C., Kumar, N., McLean, K.A., Porter, S.R., Scully,
C.M., Swan, A.V., Teo, C.G., 2000. Effect of human
immunodeciency virus-1 protease inhibitors on the clear-
ance of human herpesvirus 8 from blood of human im-
munodeciency virus-1-infected patients. J. Med. Virol. 62,
416420.
Martin, J.N., Ganem, D.E., Osmond, D.H., Page-Shafer,
K.A., Macrae, D., Kedes, D.H., 1998. Sexual transmission
and the natural history of human herpesvirus 8 infection.
New Engl. J. Med. 338, 948954.
Olsen, S.J., Chang, Y., Moore, P.S., Biggar, R.J., Melbye, M.,
1998. Increasing Kaposis sarcoma-associated herpesvirus
seroprevalence with age in a highly Kaposis sarcoma
endemic region, Zambia in 1985. AIDS 12, 19211925.
Pauk, J., Huang, M.L., Brodie, S.J., Wald, A., Koelle, D.M.,
Schacker, T., Celum, C., Selke, S., Corey, L., 2000. Mu-
cosal shedding of human herpesvirus 8 in men. New Engl.
J. Med. 343, 13691377.
Plancoulaine, S., Abel, L., van Beveren, M., Tregouet, D.A.,
Joubert, M., Tortevoye, P., de The, G., Gessain, A., 2000.
Human herpesvirus 8 transmission from mother to child
and between siblings in an endemic population. Lancet
356, 10621065.
Rainbow, L., Platt, G.M., Simpson, G.R., Sarid, R., Gao,
S.J., Stoiber, H., Herrington, C.S., Moore, P.S., Schulz,
T.F., 1997. The 222- to 234-kilodalton latent nuclear
protein (LNA) of Kaposis sarcoma-associated herpesvirus
(human herpesvirus 8) is encoded by orf73 and is a compo-
nent of the latency-associated nuclear antigen. J. Virol. 71,
59155921.
Rezza, G., Tchangmena, O.B., Andreoni, M., Bugarini, R.,
Toma, L., Bakary, D.K., Glikoutou, M., Sarmati, L.,
Monini, P., Pezzotti, P., Ensoli, B., 2000. Prevalence and
risk factors for human herpesvirus 8 infection in northern
Cameroon. Sex. Transm. Dis. 27, 159164.
Simpson, G.R., Schulz, T.F., Whitby, D., Cook, P.M.,
Boshoff, C., Rainbow, L., Howard, M.R., Gao, S.J., Bo-
henzky, R.A., Simmonds, P., Lee, C., de Ruiter, A., Hatza-
kis, A., Tedder, R.S., Weller, I.V., Weiss, R.A., Moore,
P.S., 1996. Prevalence of Kaposis sarcoma associated her-
pesvirus infection measured by antibodies to recombinant
capsid protein and latent immunouorescence antigen.
Lancet 348, 11331138.
Sitas, F., Carrara, H., Beral, V., Newton, R., Reeves, G., Bull,
D., Jentsch, U., Pacella-Norman, R., Bourboulia, D.,
Whitby, D., Boshoff, C., Weiss, R., 1999. Antibodies
against human herpesvirus 8 in black South African pa-
tients with cancer. New Engl. J. Med. 340, 18631871.
Zhang, Y.J., Deng, J.H., Rabkin, C., Gao, S.J., 2000. Hot-
spot variations of Kaposis sarcoma-associated herpesvirus
R.D.Cook et al.
/
Journal of Virological Methods
105 (2002) 297303
303
latent nuclear antigen and application in genotyping by
PCR-RFLP. J. Gen. Virol. 81, 20492058.
Zong, J.C., Ciufo, D.M., Alcendor, D.J., Wan, X., Nicholas,
J., Browning, P.J., Rady, P.L., Tyring, S.K., Orenstein,
J.M., Rabkin, C.S., Su, I.J., Powell, K.F., Croxson, M.,
Foreman, K.E., Nickoloff, B.J., Alkan, S., Hayward, G.S.,
1999. High-level variability in the ORF-K1 membrane
protein gene at the left end of the Kaposis sarcoma-associ-
ated herpesvirus genome denes four major virus subtypes
and multiple variants or clades in different human popula-
tions. J. Virol. 73, 41564170.
Zong, J., Ciufo, D.M., Viscidi, R., Alagiozoglou, L., Tyring,
S., Rady, P., Orenstein, J., Boto, W., Kalumbuja, H.,
Romano, N., Melbye, M., Kang, G.H., Boshoff, C., Hay-
ward, G.S., 2002. Genotypic analysis at multiple loci
across Kaposis sarcoma herpesvirus (KSHV) DNA
molecules: clustering patterns, novel variants and
chimerism. J. Clin. Virol. 23, 119148.
... The study group included patients with KS ( ) attending the Central Hospital of Blantyre, n p 22 Malawi, and their first-degree relatives ( ), a group of n p 67 subjects described in detail elsewhere [3, 4]. Ethical approval for the study was granted in the United Kingdom and locally. ...
... A nested polymerase chain reaction (PCR) protocol was applied to sample extracts to amplify a DNA segment from the IRD of ORF 73, as described elsewhere [4] . This segment (hereafter referred to as " IRD " ) varies in length from 1350 to 1900 bp [9]. ...
... This segment (hereafter referred to as " IRD " ) varies in length from 1350 to 1900 bp [9]. To further evaluate sequence polymorphism within IRD, PCR products were digested with BanII and MboI, after which the digested products were electrophoresed, and the gel migration distances of the products were analyzed [4]. ...
Article
Full-text available
In Malawian patients with Kaposi sarcoma (KS) and their relatives, we investigated nucleotide-sequence variation in human herpesvirus–8 (HHV-8) subgenomic DNA, amplified from oral and blood samples by use of polymerase chain reaction. Twenty-four people had amplifiable HHV-8 DNA in >1 sample; 9 (38%) were seropositive for human immunodeficiency virus type 1, 21 (88%) were anti–HHV-8–seropositive, and 7 (29%) had KS. Sequence variation was sought in 3 loci of the HHV-8 genome: the internal repeat domain of open-reading frame (ORF) 73, the KS330 segment of ORF 26, and variable region 1 of ORF K1. Significant intraperson/intersample and intrasample sequence polymorphisms were observed in 14 people (60%). For 3 patients with KS, intraperson genotypic differences, arising from nucleotide sequence variations in ORFs 26 and K1, were found in blood and oral samples. For 2 other patients with KS and for 9 people without KS, intraperson genotypic and subgenotypic differences, originating predominantly from ORF K1, were found in oral samples; for the 2 patients with KS and for 4 individuals without KS, intrasample carriage of distinct ORF K1 sequences also were discernible. Our findings imply HHV-8 superinfection
... Oropharyngeal epithelial cells were found to be a source of natural KSHV infection in immunocompetent donors, and it is likely that KSHV replicates in the oral cavity and is shed into the saliva 6 . It has also been suggested that oral transmission of KSHV occurs between healthy individuals 7,8 . KSHV causes malignancies in immunocompromised individuals, including Kaposi's sarcoma, and the virus is found primarily in endothelial cells within these tumour tissues 9 . ...
... Mediante estudios de epidemiología molecular basados en la región hipervariable del gen ORFK1, Cook y colaboradores demostraron que los diferentes miembros de la familia u otros contactos extrafamiliares fueron la fuente de infección de niños infectados con el HASK en Malawi, África. Los autores encontraron patrones de identidad y no identidad en la secuencia nucleotídica del HASK en diferentes miembros de una misma familia (17). ...
Article
Full-text available
At present, human herpesvirus or Kaposi Sarcoma-Herpes virus associated (KSHV) has been associated with the four clinical forms of Kaposi Sarcoma (classic, endemic, iatrogenic end epidemic), the primary effusion lymphoma and multicentric Castleman disease (MCD). In this review, we discuss the main hypotheses about transmission patterns of this oncogenic virus, since the pathogenic mechanisms of infection and the human to human transmission have not been fully elucidated as yet. Although Kaposi Sarcoma is not a frequent disease in Cuba, the results of studies performed by Cuban authors have demonstrated the presence of this agent in our country and its association with the variants of the epidemic and classic Kaposi Sarcoma in Cuban individuals. The knowledge of possible mechanisms involved in KSHV spread from one person to another will determine possible actions for reduction of infection by KSHV, particulary in higher risk groups.
... Human herpesvirus 8 is associated with all epidemiological variants of Kaposi's sarcoma. The mouth and oropharynx are dominant sites of viral shedding [Di Alberti et al., 1997;Koelle et al., 1997;Blackbourn et al., 1998;Lampinen et al., 2000;Pauk et al., 2000;Cook et al., 2002a;Beyari et al., 2003;Duus et al., 2004;Mbopi-Keou et al., 2004;Mbulaiteye et al., 2004;Taylor et al., 2004;Triantos et al., 2004;Widmer et al., 2006;Casper et al., 2007]. Furthermore, virus in the mouth may belong to distinct strains, reflecting multiple infection [Beyari et al., 2003]. ...
Article
Full-text available
In Saudi Arabia, the prevalence of transplantation-associated Kaposi's sarcoma (KS) is high, and there is disparity in the prevalence rates of human herpesvirus 8 (HHV-8) infection between patients with renal disease and the general population. It was hypothesized that oral HHV-8 transmission among patients undergoing hemodialysis treatment contributes to the high prevalence of infection in renal disease patients. The detection rates of anti-HHV8-IgG in plasma and HHV-8-DNA in CD45(+)-peripheral blood cells of 72 hemodialysis patients were compared first with those of 178 blood donors and 60 pregnant women. Between the hemodialysis patients and the apparently healthy people sampled, the detection rate of anti-HHV-8-IgG was 16.7% versus 0.4% (P < 0.001) and that of HHV-8-DNA was 4.2% versus 0.4%, (P < 0.05). HHV-8 DNA was determined in oral samples and the HHV-8 viral load measured in saliva of patients undergoing hemodialysis. The amount of virus shed into saliva ranged between 8,600 and 119,562,500 (mean: 24,009,360) genome-equivalents/ml among the five patients in whom oral HHV-8 DNA was detected. Finally, HHV-8-subgenomic sequencing was conducted which showed that orally shed HHV-8 in four patients belonged to genotype C2, and in one patient to genotypes A1 and C2. HHV-8 shed in the mouth of hemodialysis patients may be extensive and diverse. Oral fluid in addition to blood is thus a likely vehicle for transmission of HHV-8, possibly contributing to the high risk of HHV-8 infection in patients undergoing hemodialysis and to KS following immunosuppression after renal transplantation.
... Previous studies have detected HHV-8 DNA in the buccal mucosa and palate of KS patients (5,9,10), and in the normal oral mucosa of immunocompetent individuals (13). The present study supports these findings, but in the context of an individual who has been given immunosuppressants. ...
Article
Full-text available
Studies were conducted to investigate changes in the extent of human herpesvirus 8 (HHV-8) shedding and diversity of HHV-8 strains in the mouth of a renal allograft recipient who developed cutaneous post-transplantation Kaposi's sarcoma. Matched oral and blood samples were obtained from a Saudi Arabian renal allograft recipient from 3 days before to 38 weeks after transplantation, and from his kidney donor. Polymerase chain reaction (PCR) protocols to amplify selected HHV-8 sub-genomic regions were applied to detect and quantify HHV-8 DNA. Sequence diversity was determined by cloning the PCR products and subjecting them to denaturing gradient gel electrophoresis and to nucleotide sequencing. Before transplantation, the recipient was seropositive for anti-HHV-8 immunoglobulin G, but the donor was seronegative; HHV-8 DNA could be detected in the recipient's blood, whole-mouth saliva (WMS) and buccal exfoliates, and the salivary viral load was estimated as 2.6 million genome-copies/ml. Post-transplantation, the recipient's salivary viral load initially increased to 4.1 million genome-copies/ml, and thereafter declined precipitously, coinciding with an increase in the dosage of valaciclovir given; HHV-8 DNA was detected most often in WMS compared with parotid saliva, and buccal and palatal exfoliates. Carriage of multiple HHV-8 strains was evident in blood and oral samples; whereas before transplantation strains belonging to genotypes A1 and A5 were observed, after transplantation genotype A5 strains became dominant and A2 strains emerged. Immunosuppression and antiviral prophylaxis may interact to influence the spectrum of oral HHV-8 strains and the extent of post-transplantation HHV-8 shedding into the mouth.
Thesis
The molecular epidemiology of Kaposi's sarcoma-associated herpesvirus (KSHV) was investigated in family groups in Malawi, a region of high KSHV endemicity. It was hypothesised that: KSHV transmission may not only occur along intra-familial routes, as seroepidemiologic studies suggest, but also extra-familially; and that a host living in an endemic region may carry multiple KSHV strains. KSHV DNA in mouth rinse samples was frequently detected in study individuals (n=89 from 22 families) suggesting that saliva was a potential source of infection. Molecular sequences were compared between members of the same family by sequence analysis of hypervariable domains of opening reading frame (ORF) K1 PCR products generated from blood and oral samples. Phylogenetic analysis revealed that in some families (n=5), identical sequences in the variable region 1 were present. In others (n=4), dissimilar sequences were recovered (range of nucleotide sequence divergence: approximately 0.5% to 27%). While sequence similarity between family members is consistent with familial KSHV transmission, sequence dissimilarity and sequence clustering point to extra-familial transmission of closely related viral variants as having taken place. A PCR-based restriction fragment length polymorphism (RFLP) procedure to screen for sequence variation in the internal repeat domain of ORF 73 was then adapted to complement the ORF K1 nucleotide sequencing studies. RFLP patterns were unique for each individual and could be compared between family members. The PCR-RFLP findings broadly corroborated with the sequencing data. Intra-host KSHV sequence variation was investigated using denaturing gel gradient electrophoresis to screen multiple ORF K1 clones derived from oral samples. While intra-sample clonal diversity was observed, further investigation using KSHV infected cell lines revealed that such diversity could be attributed to Taq polymerase nucleotide misincorporation. Therefore, no firm evidence for KSHV viral diversity within single individuals could be found.
Chapter
In 1872, Moritz Kaposi, a Hungarian dermatologist, described six patients with multifocal brown-red or blue-red nodules or plaques on the feet and hands (Kaposi, 1872). Initially called “Idiopathisches multiples Pigmentsarcoma der Haut” (multiple idiopathic sarcoma of the skin) by Kaposi, the condition later became known as Kaposi's sarcoma (KS). Decades later, after the epidemiology of KS began to be investigated, its uneven geographic distribution suggested that exogenous factors were etiologically important. Subsequently, as the AIDS epidemic unfolded in the early 1980s, homosexual men were found to be up to 20 times more likely than other risk groups to develop KS, a markedly disproportionate risk that led to the hypothesis that the exogenous factor was a sexually transmitted infectious agent (Beral et al., 1990). Numerous microbial candidates were proposed (Drew et al., 1982; Huang et al., 1992; Wang et al., 1993) but for none was convincing evidence demonstrated until Kaposi's sarcoma-associated herpesvirus (KSHV), or human herpesvirus 8 (HHV-8), was discovered in 1994 (Table 54.1) (Chang et al., 1994). In a short period following the discovery of KSHV, consensus rapidly developed that it is a necessary, albeit not sufficient, causal agent of KS (Whitby et al., 1995; Gao et al., 1996a, b; Martin et al., 1998; Renwick et al.,1998; O'Brien et al., 1999). This discovery was more than academic in that, because of the AIDS epidemic, KS is now worldwide the fourth most common cancer caused by an infectious agent, following gastric, cervical, and hepatic cancer.
Article
Full-text available
Kaposi's sarcoma had been recognized as unique human cancer for a century before it manifested as an AIDS-defining illness with a suspected infectious etiology. The discovery of Kaposi's sarcoma-associated herpesvirus (KSHV), also known as human herpesvirus-8, in 1994 by using representational difference analysis, a subtractive method previously employed for cloning differences in human genomic DNA, was a fitting harbinger for the powerful bioinformatic approaches since employed to understand its pathogenesis in KS. Indeed, the discovery of KSHV was rapidly followed by publication of its complete sequence, which revealed that the virus had coopted a wide armamentarium of human genes; in the short time since then, the functions of many of these viral gene variants in cell growth control, signaling apoptosis, angiogenesis, and immunomodulation have been characterized. This critical literature review explores the pathogenic potential of these genes within the framework of current knowledge of the basic herpesvirology of KSHV, including the relationships between viral genotypic variation and the four clinicoepidemiologic forms of Kaposi's sarcoma, current viral detection methods and their utility, primary infection by KSHV, tissue culture and animal models of latent- and lytic-cycle gene expression and pathogenesis, and viral reactivation from latency. Recent advances in models of de novo endothelial infection, microarray analyses of the host response to infection, receptor identification, and cloning of full-length, infectious KSHV genomic DNA promise to reveal key molecular mechanisms of the candidate pathogeneic genes when expressed in the context of viral infection.
Article
Full-text available
Based on the observation that wild-type Kaposi's sarcoma-associated herpesvirus (KSHV) DNA can be detected in the oral cavity of healthy, immunocompetent individuals, we hypothesized that epithelial cells could be infected in vitro by wild-type (WT) KSHV isolated from immunocompetent individuals. Primary oral epithelial (P-EPI) cells and telomerase-immortalized oral epithelial cells were generated from human gingival tissue and were then infected in vitro with WT KSHV isolated from throat wash samples. Markers of lytic and latent KSHV infection were detected in cultures by 24 h postinfection by immunofluorescence confocal microscopic assays. The infectivity of the WT and BCBL virus was blocked by neutralizing antibodies against KSHV gB. The presence of KSHV DNA in these cells was confirmed by real-time PCR amplification of different regions of the viral genome. The significant in vitro viral replication that had occurred was inhibited by ganciclovir and by neutralizing antibodies against gB. When infected cultures were examined by scanning electron microscopy, thousands of KSHV particles were clearly visible across the surfaces of P-EPI cells. The detection of enveloped particles indicated that the infectious cycle had proceeded through assembly and egress. We thus demonstrated that oral WT KSHV isolated from immunocompetent individuals was able to infect and replicate in vitro in a relevant primary cell type. Furthermore, our results provide compelling evidence for KSHV transmission within infected oral epithelial cells derived from healthy, immunocompetent populations.
Article
Full-text available
Although human herpesvirus 8 (HHV-8) has been suspected to be the etiologic agent of Kaposi's sarcoma, little is known about its seroprevalence in the population, its modes of transmission, and its natural history. The San Francisco Men's Health Study, begun in 1984, is a study of a population-based sample of men in an area with a high incidence of human immunodeficiency virus (HIV) infection. We studied all 400 men infected at base line with HIV and a sample of 400 uninfected men. Base-line serum samples were assayed for antibodies to HHV-8 latency-associated nuclear antigen (anti-LANA). In addition to the seroprevalence and risk factors for anti-LANA seropositivity, we analyzed the time to the development of Kaposi's sarcoma. Anti-LANA antibodies were found in 223 of 593 men (37.6 percent) who reported any homosexual activity in the previous five years and in none of 195 exclusively heterosexual men. Anti-LANA seropositivity correlated with a history of sexually transmitted diseases and had a linear association with the number of male sexual-intercourse partners. Among the men who were infected with both HIV and HHV-8 at base line, the 10-year probability of Kaposi's sarcoma was 49.6 percent. Base-line anti-LANA seropositivity preceded and was independently associated with subsequent Kaposi's sarcoma, even after adjustment for CD4 cell counts and the number of homosexual partners. The prevalence of HHV-8 infection is high among homosexual men, correlates with the number of homosexual partners, and is temporally and independently associated with Kaposi's sarcoma. These observations are further evidence that HHV-8 has an etiologic role in Kaposi's sarcoma and is sexually transmitted among men.
Article
Full-text available
We have developed a simple, rapid, and reliable protocol for the small-scale purification of DNA and RNA from, e.g., human serum and urine. The method is based on the lysing and nuclease-inactivating properties of the chaotropic agent guanidinium thiocyanate together with the nucleic acid-binding properties of silica particles or diatoms in the presence of this agent. By using size-fractionated silica particles, nucleic acids (covalently closed circular, relaxed circular, and linear double-stranded DNA; single-stranded DNA; and rRNA) could be purified from 12 different specimens in less than 1 h and were recovered in the initial reaction vessel. Purified DNA (although significantly sheared) was a good substrate for restriction endonucleases and DNA ligase and was recovered with high yields (usually over 50%) from the picogram to the microgram level. Copurified rRNA was recovered almost undegraded. Substituting size-fractionated silica particles for diatoms (the fossilized cell walls of unicellular algae) allowed for the purification of microgram amounts of genomic DNA, plasmid DNA, and rRNA from cell-rich sources, as exemplified for pathogenic gram-negative bacteria. In this paper, we show representative experiments illustrating some characteristics of the procedure which may have wide application in clinical microbiology.
Article
Full-text available
Striking differences in Kaposi's sarcoma (KS) risk for AIDS patients who acquire HIV via homosexual activity and those whose HIV infections derive from blood product exposure suggest the presence of a sexually transmitted agent other than HIV in the development of KS. Using an immunofluorescence assay, we examined serum samples from 913 patients for the presence of antibody specific for infection by human herpesvirus 8 (HHV8), an agent whose genome is regularly found in KS tissue. The distribution of HHV8 seropositivity conforms to that expected for a sexually transmitted pathogen and tracks closely with the risk for KS development. Our data support the inference that this virus is the etiologic cofactor predicted by the epidemiology of KS.
Article
Full-text available
Kaposi's sarcoma-associated herpesvirus (KSHV), also known as human herpesvirus 8, may be the infectious cause of KS. Its prevalence in the general population, on the basis of detection of the virus genome, is controversial. To investigate the seroprevalence, we measured antibodies to a recombinant capsid-related (lytic cycle) KSHV antigen and a latent antigen complex. We selected potentially immunoreactive capsid-related proteins of KSHV by expressing them as recombinant proteins and testing them in western blot assays. We used a truncated recombinant protein encoded by KSHV open reading frame 65 (orf 65) to develop a diagnostic enzyme-linked immunosorbent assay (ELISA) and tested sera from HIV-infected individuals with KS, HIV-uninfected patients with "classic" KS, other HIV risk groups, and blood donors. We also compared the antibody response to this capsid-related protein to the response to latent antigen(s) in an immunofluorescence assay. 77/92 (84%) sera from KS patients reacted with the KSHV orf 65 protein and 84/103 (81.5%) reacted with KSHV latent antigen(s). The dominant immunogenic region of orf 65 is within the carboxyterminal 80 aminoacids, a region with little sequence similarity to the related Epstein-Barr virus, suggesting that orf 65 is a KSHV specific antigen. Only three sera from patients with haemophilia (1/84) or from intravenous drug users (2/63) had KSHV specific antibodies in the orf 65 assay whereas none of these sera reacted with latent antigen. Antibodies to KSHV were also infrequently found in UK and US blood donors by either assay (UK, 3/174 with orf 65 and 4/150 with latent antigen; US, 6/117 with orf 65 and 0/117 with latent antigen). They were more common among HIV-infected gay men without KS (5/16 by orf 65 ELISA, 10/33 by IFA), HIV-uninfected STD clinic attenders (14/166 by IFA), and Ugandan HIV-uninfected controls (6/17 by orf 65 ELISA, 9/17 by IFA). Antibody reactivity to the orf 65 protein (ELISA) and to latent antigen(s) (IFA) was concordant in 89% of 462 sera tested but reactive blood donor sera were discordant in both assays. Four AIDS-KS sera were unreactive in both assays. The distribution of antibodies to both a capsid-related recombinant protein and latent antigen(s) of KSHV strongly supports the view that infection with this virus is largely confined to individuals with, or at increased risk for, KS. However, infection with KSHV does occur, rarely, in the general UK and US population and is more common in Uganda. Antibodies to latent antigen(s) or to orf 65 encoded capsid protein will not detect all cases of KSHV infection, and a combination of several antigens will probably be required for accurate screening and confirmatory assays.
Article
Background:: The modes of transmission of HHV‐8 are still unclear. Goal:: To evaluate the distribution and transmission of HHV‐8 infection. Design:: Serosurvey conducted in a Cameroon hospital among 292 persons, including children (5‐10 years), adolescents (15‐20 years), and adults (30‐40 years). Antibodies against lytic and latent antigens to HHV‐8 were detected by immunofluorescence assay; antibodies against Epstein‐Barr virus viral antigens were detected by enzyme‐linked immunoabsorbent assay. Results:: The prevalence of HHV‐8 antilytic antibodies remained stable and was 39.8% among children, 51.5% among adolescents, and 61.8% among adults. Epstein‐Barr virus seroprevalence was high among children, and remained stable among adolescents and adults. A history of sexually transmitted diseases was an independent determinant of HHV‐8 infection (adjusted odds ratio 2.47; 95% CI 1.09‐4.91). Conclusion:: The high prevalence of HHV‐8 infection among children indicates nonsexual modes of transmission in Cameroon, with sexual transmission occurring among adolescents and adults.
Article
While in the United States and northern Europe, human herpesvirus 8 (HHV-8) appears to be mainly sexually transmitted with primary infection occurring in adulthood, the modes of transmission remain unknown in East and Central Africa, where Kaposi's sarcoma (KS) is a long-standing endemic disease, occurring not only in adults but also in children. The aim of our present study was to determine the prevalence of HHV-8 infection in children from Yaoundé, Cameroon, Central Africa. Specific antibodies directed against both latent and lytic HHV-8 antigens were detected and titrated, with an immunofluorescence assay using the KS-1 cell line, in the plasma of 258 children and adolescents, of 32 mother and child pairs and of 189 pregnant women. Two different HHV-8 DNA-specific sequences were searched in the buffy coat by PCR assays. The overall HHV-8 seroprevalence was 27.5% among these children and adolescents. In newborns, seroprevalence reached 46%, reflecting passive transmission of maternal IgG. This was followed by a marked drop. Then, beginning around 4 years of age, a regular increase of HHV-8 antibodies took place, reaching 39% in the 12- to 14-year age group and 48% above 15 years, a rate similar (54.5%) to that observed in pregnant women. PCR detection of HHV-8 sequences was negative in seronegative children and positive in the buffy coat in 17% of HHV-8-seropositive children, reflecting a low viral load in the peripheral blood. Our results establish that in Central Africa HHV-8 infection takes place during childhood by casual routes, in contrast to the sexual transmission observed in adults in northern Europe and the United States. We hypothesize that the lymphadenopathic form of KS seen in African children is related to an early and massive infection by HHV-8 in susceptible individuals. Int. J. Cancer 81:189–192, 1999. © 1999 Wiley-Liss, Inc.
Article
Background: Infection with human herpesvirus 8 (HHV-8) has been consistently linked to Kaposi's sarcoma, but its mode of transmission, association with other cancers, and interaction with the human immunodeficiency virus type 1 (HIV-1) are largely unknown. Methods: Between January 1992 and December 1997, we interviewed 3591 black patients with cancer in Johannesburg and Soweto, South Africa. Blood was tested for antibodies against HIV-1 and HHV-8 in 3344 of the patients. Antibodies against HHV-8 were detected with an indirect immunofluorescence assay. The intensity of the fluorescent signal correlated well with the titers of antibodies (P<0.001). The relations among the presence of anti-HHV-8 antibodies, sociodemographic and behavioral factors, type of cancer, and the presence or absence of coexistent HIV infection were examined with the use of unconditional logistic-regression models. Results: Among the 3293 subjects with cancers other than Kaposi's sarcoma, the standardized seroprevalence of antibodies against HHV-8 was 32 percent, which did not differ significantly from the standardized seroprevalence among black blood donors. Among these 3293 patients, the prevalence of antibodies against HHV-8 increased with increasing age (P<0.001) and an increasing number of sexual partners (P=0.05) and decreased with increasing years of education (P=0.007); it was not strongly associated with HIV-1 infection. Anti-HHV-8 antibodies were more frequent among black than white blood donors (P<0.001). Among the 51 patients with Kaposi's sarcoma, the standardized seroprevalence of antibodies against HHV-8 was 83 percent, significantly higher than the prevalence among those without Kaposi's sarcoma (P<0.001). For 16 other specific types of cancer, including multiple myeloma (108 cases) and prostate cancer (202 cases), the variation in the standardized seroprevalence of antibodies against HHV-8 was not remarkable. At a given intensity of fluorescence of anti-HHV-8 antibodies, Kaposi's sarcoma was more frequent among HIV-1-positive patients than among those who were HIV-1-negative (P<0.001). Conclusions: Among black patients with cancer in South Africa, the seroprevalence of anti-HHV-8 antibodies is high and is specifically associated with Kaposi's sarcoma, particularly at high titers.
Article
In the United States Kaposi's sarcoma is at least 20,000 times more common in persons with acquired immunodeficiency syndrome (AIDS) than in the general population and 300 times more common than in other immunosuppressed groups. Among persons with the acquired immunodeficiency syndrome (AIDS) reported to Centers for Disease Control by March 31, 1989, 15% (13,616) had Kaposi's sarcoma. Kaposi's sarcoma was commoner among those who had acquired the human immunodeficiency virus (HIV) by sexual contact than parenterally, the percentage with Kaposi's sarcoma ranging from 1% in men with haemophilia to 21% in homosexual or bisexual men. Women were more likely to have Kaposi's sarcoma if their partners were bisexual men rather than intravenous drug users. Kaposi's sarcoma risk was not consistently related to age or race but varied across the United States, being greatest in the areas that were the initial foci of the AIDS epidemic. Thus Kaposi's sarcoma in persons with AIDS may be caused by an as yet unidentified infectious agent, transmitted mainly by sexual contact.
Article
Endemic Kaposi's sarcoma (KS) is a clinically and epidemiologically distinct human immunodeficiency virus negative form of KS occurring in Africa. Kaposi's sarcoma is now the most frequently reported cancer in some areas of Africa. To determine if a KS-associated herpesvirus (KSHV) is present in both endemic HIV-seronegative and HIV-seropositive KS lesions from African patients. Paraffin-embedded tissue specimens from Ugandan patients with KS and non-KS tumor control patients attending a university-based oncology clinic were examined in a blinded case-control study. Tissue DNA specimens were examined for detectable KSHV genome by nested polymerase chain reaction performed at two independent laboratories. We identified KSHV in 17 (85%) of 20 KS tissue specimens from HIV-seronegative patients and 22 (92%) of 24 KS tissue specimens from HIV-infected persons. Kaposi's sarcoma lesions from four HIV-infected persons and four HIV-seronegative persons were positive for KSHV. Unlike previous studies in North America and Europe, three (14%) of 22 non-KS cancer control patients' tissue specimens were also positive for KSHV that resulted in an overall odds ratio of 49.2 (95% confidence interval, 9.1 to 335) for detecting KSHV in KS lesions from patients in Uganda. As in North America and Europe, KSHV infection is strongly associated with both HIV-seropositive and HIV-seronegative KS in Africa. However, it is likely that infection with this virus is more highly prevalent in Uganda.
Article
A major controversy regarding Kaposi's sarcoma-associated herpesvirus (KSHV or HHV8) is whether or not it is a ubiquitous infection of humans. Immunoassays based on KSHV- and Epstein-Barr virus (EBV)-coinfected cell lines show that most US AIDS-KS patients have specific antibodies to KSHV-related antigens. We have developed a sensitive indirect immunofluorescence assay (IFA) based on an EBV-negative, KSHV-infected cell line, BCP-1. When we used this IFA assay, KSHV-related antibodies were found in 71-88% of serum samples from US, Italian and Ugandan AIDS-KS patients, as well as all serum samples examined from HIV-seronegative KS patients. Although none of the US blood donors examined were KSHV seropositive by IFA, intermediate and high seroprevalence rates were found in Italian and Ugandan control populations. Antibody kinetics showed that more than half of the AIDS-KS patients who were examined IgG-seroconverted before KS development, and antibody levels did not decline after seroconversion. For these patients, seropositivity rates increased linearly with time, suggesting that the rate of infection was constant and that the risk of developing KS once infected with KSHV is not highly dependent on the duration of infection. These data strongly suggest that KSHV is not ubiquitous in most populations and that the virus may be under strict immunologic control in healthy KSHV-infected persons.