ArticlePDF AvailableLiterature Review

Abstract and Figures

Abstract Few years after HTLV-1 identification and isolation in humans, STLV-1, its simian counterpart, was discovered. It then became clear that STLV-1 is present almost in all simian species. Subsequent molecular epidemiology studies demonstrated that, apart from HTLV-1 subtype A, all human subtypes have a simian homolog. As HTLV-1, STLV-1 is the etiological agent of ATL, while no case of TSP/HAM has been described. Given its similarities with HTLV-1, STLV-1 represents a unique tool used for performing clinical studies, vaccine studies as well as basic science.
Content may be subject to copyright.
Jégadoetal. Retrovirology (2019) 16:41
https://doi.org/10.1186/s12977-019-0503-0
REVIEW
STLV-1 asamodel forstudying HTLV-1
infection
Brice Jégado1, Fatah Kashanchi2, Hélène Dutartre1 and Renaud Mahieux1*
Abstract
Few years after HTLV-1 identification and isolation in humans, STLV-1, its simian counterpart, was discovered. It then
became clear that STLV-1 is present almost in all simian species. Subsequent molecular epidemiology studies dem-
onstrated that, apart from HTLV-1 subtype A, all human subtypes have a simian homolog. As HTLV-1, STLV-1 is the
etiological agent of ATL, while no case of TSP/HAM has been described. Given its similarities with HTLV-1, STLV-1 repre-
sents a unique tool used for performing clinical studies, vaccine studies as well as basic science.
Keywords: HTLV-1, STLV-1, ATL, Prevalence, Interspecies transmission, Animal model, Therapy
© The Author(s) 2019. This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing,
adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and
the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material
in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material
is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the
permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creat iveco
mmons .org/licen ses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creat iveco mmons .org/publi cdoma in/
zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
Background
e first human oncogenic retrovirus was discovered in the
USA, in a T cell line obtained from blood cells of a patient
suffering from a disease then called “cutaneous T-cell lym-
phoma” [1, 2]. Few years earlier, Adult T-cell Leukemia/
Lymphoma or ATLL (i.e. an aggressive malignancy of
CD4+ T-cells) had been described in Japan [3, 4]. In 1982,
Japanese researchers also reported the presence of a ret-
rovirus among ATLL patients. ey named it Adult T cell
leukemia virus (ATLV). Further work demonstrated that
HTLV-1 specific antibodies were present among Japa-
nese ATLL patients, thus allowing identification of the first
HTLV-1 endemic area [5]. Later, it was decided to name this
virus HTLV-1 for Human T-cell Leukemia Virus type 1.
Few years later, Tropical Spastic Paraparesis/HTLV-1
associated myelopathy (TSP/HAM), a severe neuromy-
elopathy, was also identified as another disease caused
by HTLV-1 [6]. us, ATLL and TSP/HAM are the main
pathologies present among HTLV-1 infected individu-
als. It was recently estimated that 5 to 10 million people
are infected by HTLV-1 worldwide, although HTLV-1
prevalence is likely to be underestimated. Two to 4%
of HTLV-1 carriers will develop either ATLL or TSP/
HAM, while most of them will remain asymptomatic
[7]. HTLV-1 is endemic in areas such as Japan, central
Africa, the Caribbean region and South America [8].
Because HTLV-1 mostly replicates through clonal expan-
sion of infected cells even in asymptomatic carriers [9],
its retroviral genome displays a remarkable genetic stabil-
ity. HTLV-1 molecular epidemiology studies have been
carried out throughout the world. e very low genetic
variability allowed identification of different HTLV-1
subtypes. All but one of these subtypes, i.e. Cosmopoli-
tan subtype A that is present all over the world, are spe-
cific to a given African or Asian region [8]. ATL cases
were described in HTLV-1 carriers infected by HTLV-1
subtype A but also subtype B and subtype C [10, 11],
thus suggesting that ATL occurrence is not linked to the
most frequent HTLV-1 subtype. Of note, HTLV-1 sub-
type B and subtype C lack p12 and/or p30 auxiliary pro-
tein. Whether the lower ATL frequency in type B and C
infected individuals is linked to the absence of these pro-
teins remains to be determined.
In 1982, lymphocytes from a Japanese monkey
(Macaca fuscata) were co-cultured with chronically and
productively infected T-cells from the MT-2 cells, an
HTLV-1-transformed cell line. is allowed the authors
to obtain a simian cell line persistently infected by
Open Access
Retrovirology
*Correspondence: renaud.mahieux@ens-lyon.fr
1 International Center for Research in Infectiology, Retroviral Oncogenesis
Laboratory, INSERM U1111 - Université Claude Bernard Lyon 1, CNRS,
UMR5308, Ecole Normale Supérieure de Lyon, Université Lyon, Fondation
pour la Recherche Médicale, Labex Ecofect, Lyon, France
Full list of author information is available at the end of the article
Page 2 of 10
Jégadoetal. Retrovirology (2019) 16:41
HTLV-1, thus suggesting that Japanese monkeys might
be susceptible to HTLV-1 natural infection [12]. Later,
seroepidemiological studies were performed in Japan
and demonstrated that many Japanese monkeys were
infected by HTLV-1-like viruses [13]. Sera from New
World Monkeys (NWM), Old World Monkeys (OWM)
and Apes were then tested and revealed the presence
of antibodies reacting against HTLV-1 antigens. Such
antibodies were detected in OWM and Apes, but not
in NWM, suggesting endemicity of HTLV-1-related
viruses in African and Asian monkeys, but not in Ameri-
can animals [14]. Sequence analyses characterized these
viruses as Simian T-cell Leukemia Viruses (STLVs) [15,
16]. To date, it is well established that Old World Non-
Human Primates (NHPs) and Apes are naturally infected
with a great variety of STLV-1 viruses and that HTLV-1
appeared in Humans following STLV-1 cross-species
transmission approximately 27,300 years ago (95% CI
19,100–35,500) in Africa, even if interspecies transmis-
sion episodes still occur [1719]. Given the high degree
of similarity between HTLV-1 and STLV-1 sequences, it
was suggested to cluster these viruses in the single PTLV
(Primate T lymphotropic virus) family [2022]. Because
STLV-1 induces ATLL in naturally infected NHPs [23,
24], and even if some auxiliary proteins are lacking [25],
it represents a suitable tool that contributes to our under-
standing of HTLV-1 pathogenesis. is review will com-
pare HTLV-1 and STLV-1 retroviruses from different
aspects and will focus on the use of STLV-1 as a model of
HTLV-1 infection.
STLV‑1 epidemiology
Around 132 non-human primate species represent Old
World Monkeys (OWM). ey are divided in two sub-
families, Cercopithecinae and Colobinae, distributed in
African and Asian continents [26].
To determine which simian species carry STLV-1,
seroepidemiological studies were performed using kits
that had been previously developed for the detection
of anti-HTLV-1 human antibodies, as well as by PCR
(Fig. 1). Sera from Japanese monkeys were tested, and
25% scored seropositive. As in humans, STLV-1 inci-
dence increased with age and was higher in females than
males. Other species were tested later. A high seropreva-
lence was observed in African Green monkeys (AGM).
Two studies then reported STLV-1 infection in captive
Old World NHPs and Apes [27, 28]. Ishikawa etal. [29]
performed an STLV-1 survey using 567 NHPs’ blood
samples covering 30 species caught in the wild or kept
in zoos, institutes or private owners from Kenya, Gabon,
Ghana, Cameroon, Ethiopia and Indonesia. STLV-1 was
detected in African Green monkeys and Sykes’ monkeys,
in Olive baboons, Patas monkeys, Mandrills and Gorillas.
STLV-1 was also found in different species of macaques
from Indonesia, with a seroprevalence ranging from 11
to 25%. Other studies reported natural STLV-1 infec-
tions in AGM, Vervet monkeys and among baboon spe-
cies (Papio anubis, Papio hamadryas, Papio papio and
Papio cynocephalus) originating from South Africa and
Ethiopia [3033]. As in Japan, the infection status posi-
tively correlates with age, and disease incidence is higher
in females than males. Other seroepidemiological stud-
ies were also performed [3444] (Fig.1). irty-one Old
World NHP species were reported as naturally infected
with STLV-1 [33, 4550].
STLV-1 sequence analyses were then performed in
order to determine relationship between STLV-1 and
HTLV-1 and whether HTLV-1 originated from a non-
human primate virus.
STLV‑1 phylogeny
Since the first publication of a complete HTLV-1 provi-
ral genome [51], phylogenetic studies enabled to iden-
tify several HTLV-1 subtypes: Cosmopolitan subtype A,
which is found all over the world; subtypes B, D, E, F, G,
which are restricted to Central Africa; and Australo-Mel-
anesian subtype C which is the most divergent HTLV-1
subtype [8]. Based on molecular clock and phylogenetic
analyses, origin of HTLV-1 subtypes A, B, D, E was
inferred in a time frame of 27,300 ± 8200years , whereas
subtype F arose more than 10,000years ago.
In 1984, Watanabe et al. [52] demonstrated similari-
ties between restriction maps obtained using HTLV-1
from Robert Gallo’s laboratory or using Japanese simian
Adult T-cell Leukemia Virus (ATLV). ese results sug-
gested that HTLV-1 and simian ATLV shared a com-
mon ancestor. Other studies reported that HTLV-1 and
STLV-1 from Japanese monkeys, Red-faced monkeys,
Pig-tailed monkeys, AGM, Chimpanzees and baboons
(Papio cynocephalus) had the same genomic organization
i.e. LTR-gag-pol-env-pX-LTR [15, 20]. Sequence analyses
comparing Pig-tailed (Asian NHP) and AGM (African
NHP) STLV-1 sequences to HTLV-1 revealed 90% and
95% identity respectively. ese results suggested that
(1) STLV-1 could be separated into two subgroups: Asian
and African and that (2) HTLV-1 originated from the
African STLV-1 subgroup [16].
Phylogenetic studies revealed that HTLV-1 subtype B
is very closely related to STLV-1 strains infecting chim-
panzees (98% identity), Allen’s swamp monkeys (around
96% identity) and gorillas from Zaïre, Central African
Republic and Cameroon [45, 5355]. STLV-1 strains
infecting Mandrillus sphinx, Cercopithecus cephus, C.
agilis, C. pogonias, G. agilis and C. nictitans share close
relationships with HTLV-1D and -F from Cameroon
and Gabon [49, 5658]. Regarding HTLV-1 subtype E,
Page 3 of 10
Jégadoetal. Retrovirology (2019) 16:41
the Env region clusters with STLV-1 isolated from two
baboon species, Papio ursinus and Papio cynocephalus
[59]. No data has been so far reported about a simian
counterpart of HTLV-1G and HTLV-1A. Altogether,
the diversity of STLV-1 strains found in different NHPs
species and related to a given HTLV-1 subtype from the
same geographical areas is strongly supporting the con-
cept of multiple cross-species transmissions between
NHPs but also from NHPs to humans.
Most divergent STLV-1 strains were described in
Asian Macaca tonkeana (living in Indonesia) and
Macaca arctoides (living in India, ailand and China)
[6062]. Macaca tonkeana virus is related to the most
divergent HTLV-1 subtype C that is present in Melane-
sia and Australia. Molecular clock data inferred STLV-1
introduction around 156,000 to 269,000years ago on
the Asian continent [59]. ese results suggest that
macaque infection with STLV-1 might have led to the
emergence of HTLV-1 in Asian human population.
Finally, Calvignac etal. [63] demonstrated that STLV-1
sequences could be amplified from bones samples origi-
nating from an early 20th century Chlorocebus pygeryth-
rus sample. erefore, it should now be possible to use
this technique to determine STLV-1 virus evolution over
time using available Egyptian or Asian NHP mummies.
STLV‑1 interspecies transmission
Prevalence of HTLV-1 may reach 1 to 40% in adults
depending on age, sex and geographic location [8]. It is
well known that HTLV-1 can be transmitted under dif-
ferent routes: sexual, mother-to-child and contact with
infected blood. However, STLV-1 transmission occurs
mostly through aggressive contacts instead of mother
to infant or sexual transmissions [6468], even if sexual
transmission of STLV-1 is more important in NHPs such
as vervet [40].
Fig. 1 Epidemiology of Simian T-Leukemia Virus Type-1 in wild-caught or captive non-human primates (NHPs) from Asia and Africa. All studies
which reported STLV-1 infection in NHPs are listed. Orange and purple colors represent Asian and African STLV-1 infected NHPs, respectively.
Countries with both colors and hatching represent Asian and African NHPs hosted in geographical areas where they are not naturally present
Page 4 of 10
Jégadoetal. Retrovirology (2019) 16:41
STLV‑1 associated‑disease innaturally infected
animals
As it is the case for HTLV-1-infected individuals, most
STLV-1-infected monkeys remain lifelong asympto-
matic hosts [69]. For some unexplained reasons, TSP/
HAM cases have never been observed in infected
NHPs, even when those animals were living in animal
facilities for a long period. Phylogenetic studies per-
formed using samples from an African human TSP/
HAM patient showed that the viral sequence was highly
related to an STLV-1 sequence obtained from asympto-
matic West-African sooty mangabey [70]. Other strains
obtained from HTLV-1 African TSP/HAM patients also
clustered with STLV-1 strains obtained from asympto-
matic animals [71, 72]. It is well established that there is
no specific mutation in HTLV-1 genome that would be
associated with a given disease. Altogether, these data
suggest that the lack of TSP/HAM described cases in
NHPs might only be linked to the mode of viral trans-
mission rather than the age of infection.
On the contrary, a number of ATLL-like diseases
sharing clinical and pathological features with human
ATLL were reported in NHPs [24, 69, 7379]. e first
report was made in STLV-1 infected macaques which
developed malignant lymphoma [80]. Subsequent stud-
ies reported similar symptoms in captive Papio anubis,
Gorillas and AGM [7578, 81, 82]. In a recent study,
Tax-positive cells were detected in lymphoid and non-
lymphoid organs, mesenteric and axillary lymph nodes
and lung, but not in the blood from an infected Papio
anubis suffering from ATL [24]. In that case, skin lesion
biopsies also showed a massive dermal, hypodermic
and muscular cell infiltrates of positive CD3+ CD25+ T
cells, as described in human ATL.
Using STLV‑1 infected animals
After natural STLV‑1 infection
Given the high degree of sequence similarities between
STLV-1 and HTLV-1 genomes and the fact that both
viruses cause ATL, STLV-1 infected NHPs (Japanese
macaques, Mandrillus sphinx and Papio anubis) have
been used for performing molecular studies [79, 8389]
(Table1). As HTLV-1, STLV-1 infection is mostly occur-
ring in CD4+ T-cells, although STLV-1 Tax expression
was also detected in bone marrow hematopoietic stem
cells invivo, and viral DNA was retrieved in all myeloid
and lymphoid cells derived from these infected progeni-
tors [86].
STLV-1 natural infection leads to Tax and SBZ (sim-
ian equivalent of HBZ) expression. Simian SBZ and Tax
amino-acid sequences are highly similar to human HBZ
and Tax (see Tables2 and 3). ese viral proteins also
Table 1 STLV‑1 naturally or experimentally infected non‑human primates (NHPs) described in published biological
studies
STLV-1 infection mechanisms, experimental treatments and immune response were analyzed in several NHP species
Studies Natural STLV‑1 infection STLV‑1 inter‑NHPs transmission Experimental
HTLV‑1 infection
Mechanisms of (co-)infection : retroviral
replication Miura et al. [79] Dube et al. [94] Kazanji et al. [96]
Ma et al. [83] Voevodin et al. [82] Kazanji et al. [97]
Castro et al. [84] Voevodin et al. [93] Kazanji et al. [98]
Termini et al. [85] Voevodin et al. [32] Mortreux et al. [99]
Furuta et al. [86] Voevodin et al. [53] Debacq et al. [100]
Drugs and vaccine treatments Yee et al. [87] McGinn et al. [95] Heraud et al. [101]
Souquière et al. [88] Pise-Masison et al. [102]
Souquière et al. [90] Valeri et al. [103]
Souquière et al. [111] McGinn et al. [104]
Sugata et al. [89]
Cytotoxic response Turpin et al. [24]
Afonso et al. [92]
Table 2 Amino acid sequence comparison ofHTLV‑1 HBZ
vs. STLV‑1 SBZ
ATK belongs to HTLV-1 A cosmopolitan subtype, EL to HTLV-1 B subtype, STLV-1
Papio anubis was obtained from an African NHP, while STLV-1 Mf5 was obtained
from an Asian NHP (Macaca fuscata)
HTLV‑1a ATK HTLV‑1b EL
HTLV-1a ATK 74.27%
HTLV-1b EL 74.27%
STLV-1 Papio anubis 83.01% 71.36%
STLV-1 Mf5 75.71% 61.43%
Page 5 of 10
Jégadoetal. Retrovirology (2019) 16:41
display activating properties on viral LTR and NF-κB
signaling pathways. As an example, a high STLV-1 pro-
viral load (PVL) is linked to IL-2, IL-6, IL-10, IFNγ and
TNF-α elevated expression in asymptomatic STLV-
1-infected Mandrillus sphinx [90]. Given well-estab-
lished results published in the HTLV-1 situation, this
is likely due to STLV-1 Tax expression, although this
hypothesis has not been formally demonstrated. IL-2
and IFNγ results were also obtained in asymptomatic
STLV-1-positive Macaca mulatta [87], while anti IFNγ
and TNF-α responses against Tax expressing cells were
also observed in STLV-1 infected baboons [85]. STLV-1
infection also promotes CTL response against STLV-1
Tax protein [84, 85].
Interestingly, TCF1 and LEF1, two T-cell specific pro-
teins, prevent Tax effect on viral LTR. eir expression
is high in thymocytes and thus counteract STLV-1 rep-
lication in thymus. On the opposite, their expression
and thus their effect is down-regulated in peripheral
blood T-cells (both in human and simian cells), thanks
to a Tax effect on STAT5a. is might explain why Tax
is more potent in these cells, and why HTLV-1 induces
ATL in the periphery [83].
Depending upon STLV-1 strain, SBZ protein
sequence is highly similar or contain insertions and
deletion compared to HBZ (see Table2). Nevertheless,
in both cases, animals can develop ATL [24, 79]. is
might be due to conservation of the N-terminal region
as well as of C-terminus basic leucin zipper domain
between human and simian viral proteins.
As its human counterpart, STLV-1 replication occurs
through clonal expansion of infected cells, both in
asymptomatic and ATL animals [24, 79]. Antiviral ther-
apy based on the use of azidothymidine (AZT) com-
bined with interferon-α (IFN-α) improves the survival
rate of ATL patients suffering from acute and chronic/
smoldering forms. A confirmation clinical trial using
these compounds was reported in an STLV-1 infected
Papio anubis suffering from ATL. e animal was
treated with a combination of AZT and interferon-α.
However, and contrary to human ATL, no clinical
improvement was observed. It would now be interest-
ing to determine post-mortem whether, this absence of
remission was linked to p53 mutation already present
when treatment started as shown in human ATL cases
who were not responding to AZT [91].
Given the fact that treating ATL patients is difficult,
and because an elevated PVL is a characteristic of ATL,
a study tested whether PVL decreases when valproate
and AZT were delivered to asymptomatic STLV-1-in-
fected animals [92]. is was indeed the case and it was
associated to an increased anti-Tax CTL response, thus
confirming the importance of immune response for con-
trolling viral infection [92]. In another study, STLV-1
infected asymptomatic Japanese monkey were inocu-
lated with mogamulizumab (anti-CCR4), a component
that is also used for human relapsed ATL cases. is led
to a strong reduction of STLV-1 proviral load [79, 89].
Altogether, these results support the fact that STLV-1
infected animals represent a useful tool for testing drugs.
Finally, a recent study was performed in two asympto-
matic STLV-1-infected animals. is showed that immu-
nization using recombinant vaccinia viruses expressing
either Tax-22 (which cannot activate the NF-kB pathway)
or an HBZ LL/AA mutant (which is partially impaired for
blocking Tax ability to induce transcription) was linked
to a temporary decrease of STLV-1 PVL [89].
After STLV‑1 interspecies transmission
A limited number of reports described STLV-1 inter-sim-
ian species transmission [32, 53, 93, 94] (Table1). In one
report and following an unknown mode of transmission,
it was shown that baboons accidentally infected with a
rhesus macaque STLV-1 virus, developed leukemia/lym-
phoma at a high frequency [93]. is is the only reported
case suggesting that inter-simian species transmission
might impact viral pathogenesis. Experimental infection
of pig-tailed macaques with sooty mangabey STLV-1 was
also tested. Animals maintained low antibody titers and
displayed a high mortality rate without any identified
cause [95]. Finally, another work reported tantalus and
patas animals artificially infected with STLV-1 from other
species. All animals became infected, as shown by PCR
results, even if one stayed seronegative due to mutations
in the genome [94]. Why were these pol mutant viruses
still able to infect animals remains unexplained.
After articial HTLV‑1 infection
Finally, given the high degree of similarity between
HTLV-1 and STLV-1 genomes and the abundance of
molecular tools available in the HTLV-1 field, some labo-
ratories decided to use the HTLV-1 molecular clone or
HTLV-1 infected cells to perform studies in non-human
primates (Table 1). Artificial infection after inoculation
Table 3 Amino acid sequence comparison of HTLV‑1 Tax
vs. STLV‑1 Tax
ATK belongs to HTLV-1 A cosmopolitan subtype, EL to HTLV-1 B subtype, STLV-1
Papio anubis was obtained from an African NHP, while STLV-1 Mf5 was obtained
from an Asian NHP (Macaca fuscata)
HTLV‑1a ATK HTLV‑1b EL
HTLV-1a ATK 97.26%
HTLV-1b EL 97.26%
STLV-1 Papio anubis 96.03% 95.74%
STLV-1 Mf5 92.92% 93.31%
Page 6 of 10
Jégadoetal. Retrovirology (2019) 16:41
of HTLV-1 to primates provides an inestimable tool to
study primo-infection and viral dissemination, invivo, a
process that is inaccessible in humans. HTLV-1 infection
of Saimiri sciureus, i.e. non-human primates that are not
naturally infected with STLV-1 [96], demonstrated that
lymphoid organs represent the major viral reservoir [97].
As in HTLV-1 infected humans and STLV-1 naturally-
infected animals, IL-2, IL-10, IFNγ levels also increased
after HTLV-1 infection [98]. In Saimiri sciureus, the
virus also replicates through clonal expansion after hav-
ing used reverse transcription (RT) at the initial stages
[99] and it causes ATL [100]. As in baboons treated with
AZT/IFN [24], arsenic combined to IFN-α was not able
to lead to HTLV-1 proviral load reduction, even if the
number of circulating ATL flower cells decreased for
some unexplained reason [101].
Studies were also performed in pig-tailed and rhesus
macaques inoculated with autologous cells previously
transfected with the HTLV-1 ACH molecular clone
[102104]. Following infection with wild-type HTLV-
1, pig-tailed macaques developed a series of extremely
aggressive diseases that were different from ATL. ese
results therefore suggest that this animal model can-
not be used for studying events that are resulting from
HTLV-1 infection.
Consequences of rhesus macaque infection with the
same molecular clone were different since animals
remained asymptomatic. HTLV-1 p12 and p8 proteins
have been shown previously to increase NFAT activity,
IL-2 production and STAT-5 activity, while p30 controls
viral expression at the post-transcriptional level invitro
(for a review, see [105, 106]). us, this simian model was
useful to investigate the role of p12, p13, and p30 auxil-
iary proteins in vivo [102, 103]. is allowed research-
ers to show that p12 and p30 are required for allowing
HTLV-1 presence and replication in dendritic cells [103],
while p12 and p8 are necessary for allowing a viral resist-
ance to CTL responses. ese studies provided the first
invivo evidence on the mechanisms that HTLV-1 uses
to establish chronic infection and on the crucial role of
myeloid cells in that process.
Interestingly, the authors also demonstrated that the
results obtained in rhesus macaques were different from
those obtained in rabbits infected with the same viral
clones, thus reinforcing the fact that NHPs are the more
relevant system for studying HTLV-1 pathogenesis.
PTLV retroviral coinfection inNHPs andinhumans
In addition to STLV-1, other retroviruses, i.e. Simian
Immunodeficiency Virus (SIV) and Simian Foamy Virus
(SFV) infect NHPs. Cases of natural coinfection have
been reported both in humans and in NHPs: HTLV-1/
HIV-1, HTLV-1/HFV, STLV-1/SFV or STLV-1/SIV-1
[67, 107115]. HIV-1/HTLV-1 coinfection leads to sig-
nificant increase of HTLV-1 PVL as well as on a possible
delay in HIV-1 pathogenesis in humans [107, 108, 116].
Anti-HIV-1 therapy promotes an increase in HTLV-1
PVL in HIV-1/HTLV-1 coinfected carriers. ese
results strongly suggest that both retroviruses compete
for CD4+ T-cell infection. However, it is worth not-
ing that opposite results were obtained in other studies
[117121].
Natural STLV-1/SIV-1 co-infection induces the devel-
opment of a neoplastic disease in sooty mangabey [122]
and of a lymphoproliferative disease in AGM [123].
Souquière et al. described pathological manifestations,
i.e. infective dermatitis and scabies, in two STLV-1/SIV-1
co-infected mandrills [111], while no clinical sign has
been reported previously in STLV-1 naturally infected
mandrills [90]. us, these symptoms could be due to
co-infection. Ongoing experiments should allow us to
determine whether STLV-1 clonal expansion impacts SIV
replication invivo.
Finally, blood SFV proviral load from STLV-1/SFV nat-
urally co-infected Papio anubis, was recently shown to be
much higher compared to SFV mono-infected animals
[124]. ese results either suggest that cells might be co-
infected with both retroviruses, with STLV-1 promoting
clonal expansion, or that soluble STLV-1 Tax transactiva-
tor enters SFV-infected cells where it promotes viral rep-
lication. Ongoing experiments should allow us to answer
this question.
Altogether, these data demonstrate that STLV-1 is a
useful tool to understand mechanisms of HTLV-1 trans-
mission and ATL pathogenesis. PTLV-1 mono-infected
as well as SIV co-infected animals could also be used to
develop possible new anti-HTLV-1 clinical approaches
and to modify anti-HIV treatment.
Acknowledgements
BJ is supported by Labex Ecofect, RM is supported by Ecole Normale Supé-
rieure de Lyon. RM is part of the French Laboratory of Excellence project
ECOFECT (ANR-11-LABX-0048). The authors acknowledge the support Fonda-
tion pour la recherche médicale (équipe Labellisée). The authors thank Dr C.
Journo for her helpful comments.
Authors’ contributions
BJ, HD, FK and RM wrote the manuscript. All authors read and approved the
final manuscript.
Funding
RM is part of the French Laboratory of Excellence project ECOFECT (ANR-
11-LABX-0048). The authors acknowledge the support Fondation pour la
recherche médicale (équipe Labellisée DEQ20180339200). BJ is supported by
Labex Ecofect, RM is supported by Ecole Normale Supérieure de Lyon. HD is
funded by INSERM.
Availability of data and materials
Not applicable.
Ethics approval and consent to participate
Not applicable.
Page 7 of 10
Jégadoetal. Retrovirology (2019) 16:41
Consent for publication
Not applicable.
Competing interests
Not applicable.
Author details
1 International Center for Research in Infectiology, Retroviral Oncogen-
esis Laboratory, INSERM U1111 - Université Claude Bernard Lyon 1, CNRS,
UMR5308, Ecole Normale Supérieure de Lyon, Université Lyon, Fondation pour
la Recherche Médicale, Labex Ecofect, Lyon, France. 2 Laboratory of Molecular
Virology, George Mason University, Manassas, VA, USA.
Received: 24 July 2019 Accepted: 7 December 2019
References
1. Poiesz BJ, Ruscetti FW, Gazdar AF, Bunn PA, Minna JD, Gallo RC. Detec-
tion and isolation of type C retrovirus particles from fresh and cultured
lymphocytes of a patient with cutaneous T-cell lymphoma. Proc Natl
Acad Sci USA. 1980;77:7415–9.
2. Poiesz BJ, Ruscetti FW, Reitz MS, Kalyanaraman VS, Gallo RC. Isolation of
a new type C retrovirus (HTLV) in primary uncultured cells of a patient
with Sézary T-cell leukaemia. Nature. 1981;294:268–71.
3. Takatsuki K, Uchiyama T, Sagawa K. Hattori T [Lymphoma and immu-
noglobulin abnormalities, with special reference to M proteinemia].
Nippon Rinsho. 1977;35:3757–67.
4. Uchiyama T, Yodoi J, Sagawa K, Takatsuki K, Uchino H. Adult T-cell
leukemia: clinical and hematologic features of 16 cases. Blood.
1977;50:481–92.
5. Gallo RC. History of the discoveries of the first human retroviruses:
HTLV-1 and HTLV-2. Oncogene. 2005;24:5926–30.
6. Gessain A, Francis H, Sonan T, Giordano C, Akani F, Piquemal M, et al.
HTLV-I and tropical spastic paraparesis in Africa. Lancet. 1986;2:698.
7. Futsch N, Mahieux R, Dutartre H. HTLV-1, the other pathogenic yet
neglected human retrovirus: from transmission to therapeutic treat-
ment. Viruses. 2017;10:1.
8. Gessain A, Cassar O. Epidemiological aspects and world distribution of
HTLV-1 infection. Front Microbiol. 2012;3:388.
9. Wattel E, Vartanian JP, Pannetier C, Wain-Hobson S. Clonal expansion of
human T-cell leukemia virus type I-infected cells in asymptomatic and
symptomatic carriers without malignancy. J Virol. 1995;69:2863–8.
10. Mahieux R, Ibrahim F, Mauclere P, Herve V, Michel P, Tekaia F, et al.
Molecular epidemiology of 58 new African human T-cell leukemia
virus type 1 (HTLV-1) strains: identification of a new and distinct HTLV-1
molecular subtype in Central Africa and in pygmies. J Virol. 1997;71:17.
11. Einsiedel L, Cassar O, Bardy P, Kearney D, Gessain A. Variant human T-cell
lymphotropic virus type 1c and adult T-cell leukemia, Australia. Emerg
Infect Dis. 2013;19:1639–41.
12. Miyoshi I, Taguchi H, Fujishita M, Yoshimoto S, Kubonishi I, Ohtsuki Y,
et al. Transformation of monkey lymphocytes with adult T-cell leukae-
mia virus. Lancet. 1982;1:1016.
13. Miyoshi I, Yoshimoto S, Fujishita M, Taguchi H, Kubonishi I, Niiya K, et al.
Natural adult T-cell leukemia virus infection in Japanese monkeys.
Lancet. 1982;2:658.
14. Hayami M, Komuro A, Nozawa K, Shotake T, Ishikawa K, Yamamoto K,
et al. Prevalence of antibody to adult T-cell leukemia virus-associated
antigens (ATLA) in Japanese monkeys and other non-human primates.
Int J Cancer. 1984;33:179–83.
15. Komuro A, Watanabe T, Miyoshi I, Hayami M, Tsujimoto H, Seiki M, et al.
Detection and characterization of simian retroviruses homologous to
human T-cell leukemia virus type I. Virology. 1984;138:373–8.
16. Watanabe T, Seiki M, Hirayama Y, Yoshida M. Human T-cell leukemia
virus type I is a member of the African subtype of simian viruses (STLV).
Virology. 1986;148:385–8.
17. Mossoun A, Calvignac-Spencer S, Anoh AE, Pauly MS, Driscoll DA,
Michel AO, et al. Bushmeat hunting and zoonotic transmission of sim-
ian T-lymphotropic virus 1 in tropical West and Central Africa. J Virol.
2017;91:e02479-16.
18. Kazanji M, Mouinga-Ondémé A, Lekana-Douki-Etenna S, Caron M,
Makuwa M, Mahieux R, et al. Origin of HTLV-1 in hunters of nonhuman
primates in Central Africa. J Infect Dis. 2015;211:361–5.
19. Filippone C, Betsem E, Tortevoye P, Cassar O, Bassot S, Froment A,
et al. A severe bite from a nonhuman primate is a major risk factor
for HTLV-1 infection in hunters from Central Africa. Clin Infect Dis.
2015;60:1667–76.
20. Guo HG, Wong-Stall F, Gallo RC. Novel viral sequences related to human
T-cell leukemia virus in T cells of a seropositive baboon. Science.
1984;223:1195–7.
21. Watanabe T, Seiki M, Tsujimoto H, Miyoshi I, Hayami M, Yoshida M.
Sequence homology of the simian retrovirus genome with human
T-cell leukemia virus type I. Virology. 1985;144:59–65.
22. Sherman MP, Saksena NK, Dube DK, Yanagihara R, Poiesz BJ. Evolution-
ary insights on the origin of human T-cell lymphoma/leukemia virus
type I (HTLV-I) derived from sequence analysis of a new HTLV-I variant
from Papua New Guinea. J Virol. 1992;66:2556–63.
23. Locatelli S, Peeters M. Cross-species transmission of simian retroviruses:
how and why they could lead to the emergence of new diseases in the
human population. AIDS. 2012;26:659–73.
24. Turpin J, Alais S, Marçais A, Bruneau J, Melamed A, Gadot N, et al.
Whole body clonality analysis in an aggressive STLV-1 associated
leukemia (ATLL) reveals an unexpected clonal complexity. Cancer Lett.
2017;389:78–85.
25. Afonso PV, Fagrouch Z, Deijs M, Niphuis H, Bogers W, Gessain A, et al.
Absence of accessory genes in a divergent simian T-lymphotropic virus
type 1 isolated from a bonnet macaque (Macaca radiata). PLoS Negl
Trop Dis. 2019;13:e0007521.
26. Lawrence JM, Cords M. Old World Monkeys. Nat Educ Knowl.
2012;3(7):13.
27. Lowenstine LJ, Pedersen NC, Higgins J, Pallis KC, Uyeda A, Marx P, et al.
Seroepidemiologic survey of captive Old-World primates for antibodies
to human and simian retroviruses, and isolation of a lentivirus from
sooty mangabeys (Cercocebus atys). Int J Cancer. 1986;38:563–74.
28. Hunsmann G, Schneider J, Schmitt J, Yamamoto N. Detection of serum
antibodies to adult T-cell leukemia virus in non-human primates and in
people from Africa. Int J Cancer. 1983;32:329–32.
29. Ishikawa K, Fukasawa M, Tsujimoto H, Else JG, Isahakia M, Ubhi NK,
et al. Serological survey and virus isolation of simian T-cell leukemia/T-
lymphotropic virus type I (STLV-I) in non-human primates in their native
countries. Int J Cancer. 1987;40:233–9.
30. Becker WB, Becker ML, Homma T, Brede HD, Kurth R. Serum antibodies
to human T-cell leukaemia virus type I in different ethnic groups and in
non-human primates in South Africa. S Afr Med J. 1985;67:445–9.
31. Ishida T, Yamamoto K, Shotake T, Nozawa K, Hayami M, Hinuma Y. A
field study of infection with human T-cell leukemia virus among African
primates. Microbiol Immunol. 1986;30:315–21.
32. Voevodin A, Samilchuk E, Allan J, Rogers J, Broussard S. Simian
T-lymphotropic virus type 1 (STLV-1) infection in wild yellow baboons
(Papio hamadryas cynocephalus) from Mikumi National Park, Tanzania.
Virology. 1997;228:350–9.
33. Moné J, Whitehead E, Leland M, Hubbard G, Allan JS. Simian T-cell
leukemia virus type I infection in captive baboons. AIDS Res Hum
Retroviruses. 1992;8:1653–61.
34. Hayami M, Ishikawa K, Komuro A, Kawamoto Y, Nozawa K, Yamamoto
K, et al. ATLV antibody in cynomolgus monkeys in the wild. Lancet.
1983;2:620.
35. Yamamoto N, Hinuma Y, zur Hausen H, Schneider J, Hunsmann G.
African green monkeys are infected with adult T-cell leuk aemia virus or
closely related agent. Lancet. 1983;1:240–1.
36. Yamamoto N, Okada M, Hinuma Y, Hirsch FW, Chosa T, Schneider J, et al.
Human adult T-cell leukaemia virus is distinct from a similar isolate of
Japanese monkeys. J Gen Virol. 1984;65(Pt 12):2259–64.
37. Yamamoto N, Kobayashi N, Takeuchi K, Koyanagi Y, Hatanaka M, Hinuma
Y, et al. Characterization of African green monkey B-cell lines releasing
an adult T-cell leukemia-virus-related agent. Int J Cancer. 1984;34:77–82.
38. Botha MC, Jones M, de Klerk WA, Yamamoto N. Spread and distribu-
tion of human T-cell leukaemia virus type I-reactive antibody among
baboons and monkeys in the northern and eastern Transvaal. S Afr Med
J. 1985;67:665–8.
Page 8 of 10
Jégadoetal. Retrovirology (2019) 16:41
39. Coursaget P, Barres JL, Yvonnet B, Chiron JP, Cornet M, Ferrara L, et al.
Antibodies to human T-cell leukemia virus (HTLV-1) in non human
primates from Senegal. Biomed Pharmacother. 1985;39:198–9.
40. Dracopoli NC, Turner TR, Else JG, Jolly CJ, Anthony R, Gallo RC, et al.
STLV-I antibodies in feral populations of East African vervet monkeys
(Cercopithecus aethiops). Int J Cancer. 1986;38:523–9.
41. Daniel MD, Letvin NL, Sehgal PK, Schmidt DK, Silva DP, Solomon KR,
et al. Prevalence of antibodies to 3 retroviruses in a captive colony of
macaque monkeys. Int J Cancer. 1988;41:601–8.
42. Fultz PN, Gordon TP, Anderson DC, McClure HM. Prevalence of natural
infection with simian immunodeficiency virus and simian T-cell leu-
kemia virus type I in a breeding colony of sooty mangabey monkeys.
AIDS. 1990;4:619–25.
43. Estaquier J, Peeters M, Bedjabaga L, Honoré C, Bussi P, Dixson A, et al.
Prevalence and transmission of simian immunodeficiency virus and
simian T-cell leukemia virus in a semi-free-range breeding colony of
mandrills in Gabon. AIDS. 1991;5:1385–6.
44. Courgnaud V, Van Dooren S, Liegeois F, Pourrut X, Abela B, Loul S, et al.
Simian T-cell leukemia virus (STLV ) infection in wild primate popula-
tions in Cameroon: evidence for dual STLV type 1 and type 3 infection
in agile mangabeys (Cercocebus agilis). J Virol. 2004;78:4700–9.
45. Koralnik IJ, Boeri E, Saxinger WC, Monico AL, Fullen J, Gessain A, et al.
Phylogenetic associations of human and simian T-cell leukemia/lym-
photropic virus type I strains: evidence for interspecies transmission. J
Virol. 1994;68:2693–707.
46. Mahieux R, Pecon-Slattery J, Chen GM, Gessain A. Evolutionary infer-
ences of novel simian T lymphotropic virus type 1 from Wild-Caught
Chacma (Papio ursinus) and olive baboons (Papio anubis). Virology.
1998;251:71–84.
47. Saksena NK, Her ve V, Durand JP, Leguenno B, Diop OM, Digouette
JP, et al. Seroepidemiologic, molecular, and phylogenetic analyses
of simian T-cell leukemia viruses (STLV-I) from various naturally
infected monkey species from central and western Africa. Virology.
1994;198:297–310.
48. Schatzl H, Yakovleva L, Lapin B, Rose D, Inzhiia L, Gaedigk-Nitschko
K, et al. Detection and characterization of T-cell leukemia virus-like
proviral sequences in PBL and tissues of baboons by PCR. Leukemia.
1992;6(Suppl 3):158S–60S.
49. Sintasath DM, Wolfe ND, LeBreton M, Jia H, Garcia AD, Diffo JLD, et al.
Simian T-lymphotropic virus diversity among nonhuman primates,
Cameroon. Emerg Infect Dis. 2009;15:175–84.
50. Van Dooren S, Verschoor EJ, Fagrouch Z, Vandamme A-M. Phylogeny
of primate T lymphotropic virus type 1 (PTLV-1) including various
new Asian and African non-human primate strains. Infect Genet Evol.
2007;7:374–81.
51. Seiki M, Hattori S, Hirayama Y, Yoshida M. Human adult T-cell leukemia
virus: complete nucleotide sequence of the provirus genome inte-
grated in leukemia cell DNA. Proc Natl Acad Sci USA. 1983;80:3618–22.
52. Watanabe T, Seiki M, Yoshida M. HTLV type I (U. S. isolate) and ATLV
(Japanese isolate) are the same species of human retrovirus. Virology.
1984;133:238–41.
53. Voevodin AF, Johnson BK, Samilchuk EI, Stone GA, Druilhet R, Greer WJ,
et al. Phylogenetic analysis of simian T-lymphotropic virus type I (STLV-I)
in common chimpanzees (Pan troglodytes): evidence for interspecies
transmission of the virus between chimpanzees and humans in Central
Africa. Virology. 1997;238:212–20.
54. Meertens L, Rigoulet J, Mauclère P, Van Beveren M, Chen GM, Diop
O, et al. Molecular and phylogenetic analyses of 16 novel simian T
cell leukemia virus type 1 from Africa: close relationship of STLV-1
from Allenopithecus nigroviridis to HTLV-1 subtype B strains. Virology.
2001;287:275–85.
55. Ayouba A, Michem A, Peeters M, Vercammen F. Full-genome characteri-
zation of simian T-cell leukemia virus type 1 subtype b from a wild-born
captive gorilla gorilla gorilla with T-cell lymphoma. Genome Announc.
2017;5:e01117-17.
56. Mahieux R, Chappey C, Georges-Courbot MC, Dubreuil G, Mauclere P,
Georges A, et al. Simian T-cell lymphotropic virus type 1 from Mandrillus
sphinx as a simian counterpart of human T-cell lymphotropic virus type
1 subtype D. J Virol. 1998;72:10316–22.
57. Makuwa M, Souquière S, Clifford SL, Telfer PT, Sallé B, Bourry O, et al.
Two distinct STLV-1 subtypes infecting Mandrillus sphinx follow the
geographic distribution of their hosts. AIDS Res Hum Retroviruses.
2004;20:1137–43.
58. Liégeois F, Lafay B, Switzer WM, Locatelli S, Mpoudi-Ngolé E, Loul S, et al.
Identification and molecular characterization of new STLV-1 and STLV-3
strains in wild-caught nonhuman primates in Cameroon. Virology.
2008;371:405–17.
59. Van Dooren S, Salemi M, Vandamme AM. Dating the origin of the Afri-
can human T-cell lymphotropic virus type-i (HTLV-I) subtypes. Mol Biol
Evol. 2001;18:661–71.
60. Mahieux R, Pecon-Slattery J, Gessain A. Molecular characterization
and phylogenetic analyses of a new, highly divergent simian T-cell
lymphotropic virus type 1 (STLV-1marc1) in Macaca arctoides. J Virol.
1997;71:6253–8.
61. Ibrahim F, de Thé G, Gessain A. Isolation and characterization of a new
simian T-cell leukemia virus type 1 from naturally infected celebes
macaques (Macaca tonkeana): complete nucleotide sequence and
phylogenetic relationship with the Australo-Melanesian human T-cell
leukemia virus type 1. J Virol. 1995;69:6980–93.
62. Van Dooren S, Meertens L, Lemey P, Gessain A, Vandamme A-M. Full-
genome analysis of a highly divergent simian T-cell lymphotropic virus
type 1 strain in Macaca arctoides. J Gen Virol. 2005;86:1953–9.
63. Calvignac S, Terme J-M, Hensley SM, Jalinot P, Greenwood AD, Hänni C.
Ancient DNA identification of early 20th century simian T-cell leukemia
virus type 1. Mol Biol Evol. 2008;25:1093–8.
64. Niphuis H, Verschoor EJ, Bontjer I, Peeters M, Heeney JL. Reduced trans-
mission and prevalence of simian T-cell lymphotropic virus in a closed
breeding colony of chimpanzees (Pan troglodytes verus). J Gen Virol.
2003;84:615–20.
65. Leendertz FH, Junglen S, Boesch C, Formenty P, Couacy-Hymann E,
Courgnaud V, et al. High variety of different simian T-cell leukemia virus
type 1 strains in chimpanzees (Pan troglodytes verus) of the Taï National
Park, Côte d’Ivoire. J Virol. 2004;78:4352–6.
66. d’Offay JM, Eberle R, Sucol Y, Schoelkopf L, White MA, Valentine BD, et al.
Transmission dynamics of simian T-lymphotropic virus type 1 (STLV1)
in a baboon breeding colony: predominance of female-to-female trans-
mission. Comp Med. 2007;57:105–14.
67. Nerrienet E, Amouretti X, Müller-Trutwin MC, Poaty-Mavoungou V, Bed-
jebaga I, Nguyen HT, et al. Phylogenetic analysis of SIV and STLV type I
in mandrills (Mandrillus sphinx): indications that intracolony transmis-
sions are predominantly the result of male-to-male aggressive contacts.
AIDS Res Hum Retroviruses. 1998;14:785–96.
68. Roussel M, Pontier D, Ngoubangoye B, Kazanji M, Verrier D, Fouchet D.
Modes of transmission of simian T-lymphotropic virus type 1 in semi-
captive mandrills (Mandrillus sphinx). Vet Microbiol. 2015;179:155–61.
69. Allan JS, Leland M, Broussard S, Mone J, Hubbard G. Simian T-cell
lymphotropic viruses (STLVs) and lymphomas in african nonhuman
primates. Cancer Invest. 2001;19:383–95.
70. Enose-Akahata Y, Caruso B, Haner B, Charlip E, Nair G, Massoud R, et al.
Development of neurologic diseases in a patient with primate T lym-
photropic virus type 1 (PTLV-1). Retrovirology. 2016;13:56.
71. Salemi M, Van Dooren S, Audenaert E, Delaporte E, Goubau P, Desmyter
J, et al. Two new human T-lymphotropic virus type I phylogenetic
subtypes in seroindeterminates, a Mbuti pygmy and a Gabonese, have
closest relatives among African STLV-I strains. Virology. 1998;246:277–87.
72. Touzé E, Gessain A, Lyon-Caen O, Gout O. Tropical spastic paraparesis/
HTLV-I-associated myelopathy in Europe and in Africa: clinical and
epidemiologic aspects. J Acquir Immune Defic Syndr Hum Retrovirol.
1996;13(Suppl 1):S38–45.
73. Noda Y, Ishikawa K, Sasagawa A, Honjo S, Mori S, Tsujimoto H, et al.
Hematologic abnormalities similar to the preleukemic state of adult
T-cell leukemia in African green monkeys naturally infected with simian
T-cell leukemia virus. Jpn J Cancer Res. 1986;77:1227–34.
74. Sakakibara I, Sugimoto Y, Sasagawa A, Honjo S, Tsujimoto H, Nakamura
H, et al. Spontaneous malignant lymphoma in an African green monkey
naturally infected with simian T-lymphotropic virus (STLV). J Med Prima-
tol. 1986;15:311–8.
75. Tsujimoto H, Noda Y, Ishikawa K, Nakamura H, Fukasawa M, Sakak-
ibara I, et al. Development of adult T-cell leukemia-like disease in
African green monkey associated with clonal integration of simian
T-cell leukemia virus type I. Cancer Res. 1987;47:269–74.
Page 9 of 10
Jégadoetal. Retrovirology (2019) 16:41
76. McCarthy TJ, Kennedy JL, Blakeslee JR, Bennett BT. Spontaneous
malignant lymphoma and leukemia in a simian T-lymphotropic
virus type I (STLV-I) antibody positive olive baboon. Lab Anim Sci.
1990;40:79–81.
77. Hubbard GB, Moné JP, Allan JS, Davis KJ, Leland MM, Banks PM, et al.
Spontaneously generated non-Hodgkin’s lymphoma in twenty-seven
simian T-cell leukemia virus type 1 antibody-positive baboons (Papio
species). Lab Anim Sci. 1993;43:301–9.
78. d’Offay JM, Eberle R, Wolf RF, Kosanke SD, Doocy KR, Ayalew S, et al.
Simian T-lymphotropic Virus-associated lymphoma in 2 naturally
infected baboons: T-cell clonal expansion and immune response dur-
ing tumor development. Comp Med. 2013;63:288–94.
79. Miura M, Yasunaga J, Tanabe J, Sugata K, Zhao T, Ma G, et al. Charac-
terization of simian T-cell leukemia virus type 1 in naturally infected
Japanese macaques as a model of HTLV-1 infection. Retrovirology.
2013;10:118.
80. Homma T, Kanki PJ, King NW, Hunt RD, O’Connell MJ, Letvin NL, et al.
Lymphoma in macaques: association with virus of human T lympho-
trophic family. Science. 1984;225:716–8.
81. Srivastava BI, Wong-Staal F, Getchell JP. Human T-cell leukemia virus
I provirus and antibodies in a captive gorilla with non-Hodgkin’s
lymphoma. Cancer Res. 1986;46:4756–8.
82. Voevodin AF, Lapin BA, Yakovleva LA, Ponomaryeva TI, Oganyan
TE, Razmadze EN. Antibodies reacting with human T-lymphotropic
retrovirus (HTLV-I) or related antigens in lymphomatous and healthy
hamadryas baboons. Int J Cancer. 1985;36:579–84.
83. Ma G, Yasunaga J, Akari H, Matsuoka M. TCF1 and LEF1 act as T-cell
intrinsic HTLV-1 antagonists by targeting Tax. Proc Natl Acad Sci USA.
2015;112:2216–21.
84. Castro I, Giret TM, Magnani DM, Maxwell HS, Umland O, Perry JK, et al.
Cellular immune responses against simian T-lymphotropic virus type
1 target tax in infected baboons. J Virol. 2016;90:5280–91.
85. Termini JM, Magnani DM, Maxwell HS, Lauer W, Castro I, Pecotte
J, et al. Simian T lymphotropic virus 1 infection of Papio anu-
bis: tax sequence heterogeneity and T cell recognition. J Virol.
2017;91:e00950-17.
86. Furuta R, Yasunaga J-I, Miura M, Sugata K, Saito A, Akari H, et al. Human
T-cell leukemia virus type 1 infects multiple lineage hematopoietic cells
in vivo. PLoS Pathog. 2017;13:e1006722.
87. Yee JL, Montiel NA, Ardeshir A, Ardeshr A, Lerche NW. Constitutive
release of IFNγ and IL2 from peripheral blood mononuclear cells of rhe-
sus macaques (Macaca mulatta) infected with simian T-lymphotropic
virus type 1. Comp Med. 2013;63:508–14.
88. Souquière S, Mouinga-Ondemé A, Makuwa M, Hermine O, Kazanji M.
Dynamic interaction between STLV-1 proviral load and T-cell response
during chronic infection and after immunosuppression in non-human
primates. PLoS ONE. 2009;4:e6050.
89. Sugata K, Yasunaga J-I, Miura M, Akari H, Utsunomiya A, Nosaka K,
et al. Enhancement of anti-STLV-1/HTLV-1 immune responses through
multimodal effects of anti-CCR4 antibody. Sci Rep. 2016;6:27150.
90. Souquière S, Mouinga-Ondeme A, Makuwa M, Beggio P, Radaelli A, De
Giuli Morghen C, et al. T-cell tropism of simian T-cell leukaemia virus
type 1 and cytokine profiles in relation to proviral load and immuno-
logical changes during chronic infection of naturally infected mandrills
(Mandrillus sphinx). J Med Primatol. 2009;38:279–89.
91. Datta A, Bellon M, Sinha-Datta U, Bazarbachi A, Lepelletier Y, Canioni D,
et al. Persistent inhibition of telomerase reprograms adult T-cell leuke-
mia to p53-dependent senescence. Blood. 2006;108:1021–9.
92. Afonso PV, Mekaouche M, Mortreux F, Toulza F, Moriceau A, Wattel
E, et al. Highly active antiretroviral treatment against STLV-1 infec-
tion combining reverse transcriptase and HDAC inhibitors. Blood.
2010;116:3802–8.
93. Voevodin A, Samilchuk E, Schätzl H, Boeri E, Franchini G. Interspecies
transmission of macaque simian T-cell leukemia/lymphoma virus type
1 in baboons resulted in an outbreak of malignant lymphoma. J Virol.
1996;70:1633–9.
94. Dube S, Saksena N, Spicer T, Healey J, Benz P, Dube DK, et al. Delayed
seroconversion to STLV-1 infection is associated with mutations in the
pol and rex genes. Virol J. 2013;10:282.
95. McGinn TM, Tao B, Cartner S, Schoeb T, Davis I, Ratner L, et al. Asso-
ciation of primate T-cell lymphotropic virus infection of pig-tailed
macaques with high mortality. Virology. 2002;304:364–78.
96. Kazanji M, Moreau JP, Mahieux R, Bonnemains B, Bomford R, Gessain
A, et al. HTLV-I infection in squirrel monkeys (Saïmiri sciureus) using
autologous, homologous, or heterologous HTLV-I-transformed cell
lines. Virology. 1997;231:258–66.
97. Kazanji M, Ureta-Vidal A, Ozden S, Tangy F, de Thoisy B, Fiette L, et al.
Lymphoid organs as a major reservoir for human T-cell leukemia virus
type 1 in experimentally infected squirrel monkeys (Saimiri sciureus):
provirus expression, persistence, and humoral and cellular immune
responses. J Virol. 2000;74:4860–7.
98. Kazanji M, Heraud J-M, Merien F, Pique C, de Thé G, Gessain A, et al.
Chimeric peptide vaccine composed of B- and T-cell epitopes of
human T-cell leukemia virus type 1 induces humoral and cellular
immune responses and reduces the proviral load in immunized squirrel
monkeys (Saimiri sciureus). J Gen Virol. 2006;87:1331–7.
99. Mortreux F, Kazanji M, Gabet AS, de Thoisy B, Wattel E. Two-step nature
of human T-cell leukemia virus type 1 replication in experimentally
infected squirrel monkeys (Saimiri sciureus). J Virol. 2001;75:1083–9.
100. Debacq C, Héraud J-M, Asquith B, Bangham C, Merien F, Moules V, et al.
Reduced cell turnover in lymphocytic monkeys infected by human
T-lymphotropic virus type 1. Oncogene. 2005;24:7514–23.
101. Heraud JM, Mortreux F, Merien F, Contamin H, Mahieux R, Pouliquen
JF, et al. The efficacy of combined therapy of arsenic trioxide and alpha
interferon in human T-cell leukemia virus type-1-infected squirrel
monkeys (Saimiri sciureus). Antiviral Res. 2006;70:132–9.
102. Pise-Masison CA, de Castro-Amarante MF, Enose-Akahata Y, Buchmann
RC, Fenizia C, Washington Parks R, et al. Co-dependence of HTLV-1 p12
and p8 functions in virus persistence. PLoS Pathog. 2014;10:e1004454.
103. Valeri VW, Hryniewicz A, Andresen V, Jones K, Fenizia C, Bialuk I, et al.
Requirement of the human T-cell leukemia virus p12 and p30 products
for infectivity of human dendritic cells and macaques but not rabbits.
Blood. 2010;116:3809–17.
104. McGinn TM, Wei Q, Stallworth J, Fultz PN. Immune responses to HTLV-
I(ACH) during acute infection of pig-tailed macaques. AIDS Res Hum
Retroviruses. 2004;20:443–56.
105. Bai XT, Nicot C. Overview on HTLV-1 p12, p8, p30, p13: accomplices in
persistent infection and viral pathogenesis. Front Microbiol. 2012;3:400.
106. Anupam R, Doueiri R, Green PL. The need to accessorize: molecular
roles of HTLV-1 p30 and HTLV-2 p28 accessory proteins in the viral life
cycle. Front Microbiol. 2013;4:275.
107. Beilke MA, Theall KP, O’Brien M, Clayton JL, Benjamin SM, Winsor EL,
et al. Clinical outcomes and disease progression among patients coin-
fected with HIV and human T lymphotropic virus types 1 and 2. Clin
Infect Dis. 2004;39:256–63.
108. Oo Z, Barrios CS, Castillo L, Beilke MA. High levels of CC-chemokine
expression and downregulated levels of CCR5 during HIV-1/HTLV-1 and
HIV-1/HTLV-2 coinfections: CC-chemokines and CCR5 levels during HIV/
HTLV coinfections. J Med Virol. 2015;87:790–7.
109. Switzer WM, Garcia AD, Yang C, Wright A, Kalish ML, Folks TM, et al.
Coinfection with HIV-1 and simian foamy virus in West Central Africans.
J Infect Dis. 2008;197:1389–93.
110. Switzer WM, Tang S, Zheng H, Shankar A, Sprinkle PS, Sullivan V, et al.
Dual simian foamy virus/human immunodeficiency virus type 1 infec-
tions in persons from Côte d’Ivoire. PLoS ONE. 2016;11:e0157709.
111. Souquière S, Makuwa M, Sallé B, Lepelletier Y, Mortreux F, Hermine O,
et al. Immunological alterations and associated diseases in mandrills
(Mandrillus sphinx) naturally co-infected with SIV and STLV. Virology.
2014;454–455:184–96.
112. Traina-Dorge VL, Martin LN, Lorino R, Winsor EL, Beilke MA. Human
T cell leukemia virus type 1 up-regulation after simian immunode-
ficiency virus-1 coinfection in the nonhuman primate. J Infect Dis.
2007;195:562–71.
113. Harrison LH, Quinn TC, Schechter M. Human T cell lymphotropic virus
type I does not increase human immunodeficiency virus viral load
in vivo. J Infect Dis. 1997;175:438–40.
114. Goldberg TL, Sintasath DM, Chapman CA, Cameron KM, Karesh WB,
Tang S, et al. Coinfection of ugandan red colobus (Procolobus [Piliocolo-
bus] rufomitratus tephrosceles) with novel, divergent delta-, lenti-, and
spumaretroviruses. J Virol. 2009;83:11318–29.
Page 10 of 10
Jégadoetal. Retrovirology (2019) 16:41
fast, convenient online submission
thorough peer review by experienced researchers in your field
rapid publication on acceptance
support for research data, including large and complex data types
gold Open Access which fosters wider collaboration and increased citations
maximum visibility for your research: over 100M website views per year
At BMC, research is always in progress.
Learn more biomedcentral.com/submissions
Ready to submit your research
? Choose BMC and benefit from:
115. Durand JP, Tuppin P, Maison P, Galat G, Galat-Luong A, Jeannel D, et al.
Increased risk for a second retroviral infection (SIV or STLV type I) for
wild African green monkeys already infected by one retrovirus in Sen-
egal (West Africa). AIDS Res Hum Retroviruses. 1995;11:985–8.
116. Beilke MA, Dorge VLT, Sirois M, Bhuiyan A, Murphy EL, Walls JM, et al.
Relationship between human T lymphotropic virus (HTLV ) type 1/2 viral
burden and clinical and treatment parameters among patients with HIV
type 1 and HTLV-1/2 coinfection. Clin Infect Dis. 2007;44:1229–34.
117. Beilke MA, Japa S, Moeller-Hadi C, Martin-Schild S. Tropical spastic
paraparesis/human T leukemia virus type 1-associated myelopathy in
HIV type 1-coinfected patients. Clin Infect Dis. 2005;41:e57–63.
118. Casseb J, Posada-Vergara MP, Montanheiro P, Fukumori LMI, Olah I, Smid
J, et al. T CD4+ cells count among patients co-infected with human
immunodeficiency virus type 1 (HIV-1) and human T-cell leukemia virus
type 1 (HTLV-1): high prevalence of tropical spastic paraparesis/HTLV-
1-associated myelopathy (TSP/HAM). Revista do Instituto de Medicina
Tropical de São Paulo. 2007;49:231–3.
119. Casseb J, de Oliveira ACP, Vergara MPP, Montanheiro P, Bonasser F, Meil-
man Ferreira C, et al. Presence of tropical spastic paraparesis/human
T-cell lymphotropic virus type 1-associated myelopathy (TSP/HAM)-like
among HIV-1-infected patients. J Med Virol. 2008;80:392–8.
120. Casoli C, Pilotti E, Bertazzoni U. Molecular and cellular interactions of
HIV-1/HTLV coinfection and impact on AIDS progression. AIDS Rev.
2007;9:140–9.
121. Rahimi H, Rezaee SA, Valizade N, Vakili R, Rafatpanah H. Assessment
of HTLV-I proviral load, HIV viral load and CD4 T cell count in infected
subjects; with an emphasis on viral replication in co-infection. Iran J
Basic Med Sci. 2014;17:49–54.
122. Fultz PN, Su L, May P, West JT. Isolation of sooty mangabey simian T-cell
leukemia virus type I [STLV-I(sm)] and characterization of a mangabey
T-cell line coinfected with STLV-I(sm) and simian immunodeficiency
virus SIVsmmPBj14. Virology. 1997;235:271–85.
123. Traina-Dorge V, Blanchard J, Martin L, Murphey-Corb M. Immunode-
ficiency and lymphoproliferative disease in an African green monkey
dually infected with SIV and STLV-I. AIDS Res Hum Retroviruses.
1992;8:97–100.
124. Alais S, Pasquier A, Jegado B, Journo C, Rua R, Gessain A, et al. STLV-1
co-infection is correlated with an increased SFV proviral load in the
peripheral blood of SFV/STLV-1 naturally infected non-human primates.
PLoS Negl Trop Dis. 2018;12:e0006812.
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in pub-
lished maps and institutional affiliations.
... Various animal models have been used to elucidate the pathogenesis of ATL and develop relevant therapies. Old World monkeys and apes, including Japanese monkeys and Cynomolgus monkeys, are naturally infected with STLV-1, which is closely related to HTLV-1 [24]. STLV-1-infected monkey models have been used to study vaccines and therapeutics for ATL [25][26][27][28]. ...
Article
Full-text available
Adult T cell leukemia (ATL) is an aggressive hematologic disease caused by human T cell leukemia virus type 1 (HTLV-1) infection. Various animal models of HTLV-1 infection/ATL have been established to elucidate the pathogenesis of ATL and develop appropriate treatments. For analyses employing murine models, transgenic and immunodeficient mice are used because of the low infectivity of HTLV-1 in mice. Each mouse model has different characteristics that must be considered before use for different HTLV-1 research purposes. HTLV-1 Tax and HBZ transgenic mice spontaneously develop tumors, and the roles of both Tax and HBZ in cell transformation and tumor growth have been established. Severely immunodeficient mice were able to be engrafted with ATL cell lines and have been used in preclinical studies of candidate molecules for the treatment of ATL. HTLV-1-infected humanized mice with an established human immune system are a suitable model to characterize cells in the early stages of HTLV-1 infection. This review outlines the characteristics of mouse models of HTLV-1 infection/ATL and describes progress made in elucidating the pathogenesis of ATL and developing related therapies using these mice.
... HTLV, in particular HTLV-1 and HTLV-3, appeared by cross-species transmission from monkeys to humans, which is a process still ongoing in Africa [189,194]. Based on the cross-species transmission, a high similarity of the HTLV-1/HTLV-3 and STLV-1/STLV-3 genome is given, e.g., 96% of the Tax sequence is identical between HTLV-1a ATK and STLV-1 Papio anubis strain [195,196]. While the STLV-2 strain has only been reported in wild-born bonobos in captivity, STLV-4 has been detected with PCR in gorillas in Cameroon and seems to be endemic [197]. ...
Article
Full-text available
The transmission of viruses from one host to another typically occurs through horizontal or vertical pathways. The horizontal pathways include transmission amongst individuals, usually through bodily fluids or excretions, while vertical transmission transpires from mother to their offspring, either during pregnancy, childbirth, or breastfeeding. While there are more than 200 human pathogenic viruses to date, only a small number of them are known to be transmitted via breast milk, including cytomegalovirus (CMV), human immunodeficiency virus type 1 (HIV-1), and human T cell lymphotropic virus type 1 (HTLV-1), the latter two belonging to the family Retroviridae. Breast milk transmission is a common characteristic among mammalian retroviruses, but there is a lack of reports summarizing our knowledge regarding this route of transmission of mammalian retroviruses. Here, we provide an overview of the transmission of mammalian exogenous retroviruses with a focus on Orthoretrovirinae, and we highlight whether they have been described or suspected to be transmitted through breast milk, covering various species. We also elaborate on the production and composition of breast milk and discuss potential entry sites of exogenous mammalian retroviruses during oral transmission.
... Notably, investigating HTLV-1 neurotropism is a challenging task due to (i) the absence of suitable in vitro models that can recapitulate the complexity of the Central Nervous System (CNS) cytoarchitecture, (ii) the limited access to myelinated postmortem samples from infected individuals, and (iii) the lack of tools that are available to be used to investigate HTLV-1 infections (i.e., GFP-reportable viral particles), compared to infections by other retroviruses. In contrast, naturally STLV-1-infected nonhuman primates (NHP) represent a valuable animal model for HLTV-1 infection, as the simian homolog of HTLV-1 shares more than 99% of its genomic homology (12) and offers the possibility of accessing neural tissue before the demyelinating phase of the infection. ...
Article
Full-text available
The human T-cell leukemia virus (HTLV)-1 is responsible for an aggressive neurodegenerative disease (HAM/TSP) and multiple neurological alterations. The capacity of HTLV-1 to infect central nervous system (CNS) resident cells, together with the neuroimmune-driven response, has not been well-established. Here, we combined the use of human induced pluripotent stem cells (hiPSC) and of naturally STLV-1-infected nonhuman primates (NHP) as models with which to investigate HTLV-1 neurotropism. Hence, neuronal cells obtained after hiPSC differentiation in neural polycultures were the main cell population infected by HTLV-1. Further, we report the infection of neurons with STLV-1 in spinal cord regions as well as in brain cortical and cerebellar sections of postmortem NHP. Additionally, reactive microglial cells were found in infected areas, suggesting an immune antiviral response. These results emphasize the need to develop new efficient models by which to understand HTLV-1 neuroinfection and suggest an alternative mechanism that leads to HAM/TSP.
... Therefore, these observations suggest that this region is likely to be the center of origin of deltaretroviruses (Slattery et al., 1999;Reid et al., 2016;Afonso et al., 2019). Further analysis is needed to detect other natural hosts of BLV, because the infection rate in zebu cattle is relatively low for the maintenance of deltaretrovirus as a reservoir (Mwenda et al., 1999;Takemura et al., 2002;Traina-Dorge et al., 2005;Jegado et al., 2019). ...
Article
Full-text available
Bovine leukemia virus (BLV), the causative agent of enzootic bovine leukosis, is currently one of the most important pathogens affecting the cattle industry worldwide. Determining where and in which host it originated, and how it dispersed across continents will provide valuable insights into its historical emergence as the cattle pathogen. Various species in the Bos genus were domesticated in Asia, where they also diversified. As native cattle (taurine cattle, zebu cattle, yak, and water buffalo) are indigenous and adapted to local environments, we hypothesized that Asian native cattle could have harbored BLV and, therefore, that they were important for virus emergence, maintenance, and spread. In this study, phylogeographic and ancestral trait analyses—including sequences obtained from Asian native cattle—were used to reconstruct the evolutionary history of BLV. It was shown that, since its probable emergence in Asia, the virus spread to South America and Europe via international trade of live cattle. It was inferred that zebu cattle were the hosts for the early origin of BLV, while taurine cattle played the significant role in the transmission worldwide. In addition, the results of positive selection analysis indicate that yak had a substantially minor role in the transmission of this virus. In this study, endogenous deltaretrovirus sequences in bats, collected in Asian countries, were also analyzed on whether these sequences were present in the bat genome. Endogenous deltaretrovirus sequences were detected from bat species endemic to specific regions and geographically isolated for a long time. Endogenous deltaretrovirus sequences from these geographically isolated species represent ancient exogenous deltaretroviruses distributions. The phylogenetic analysis revealed that these newly obtained endogenous deltaretrovirus sequences were closely related to those of BLV from Asian native cattle, indicating that BLV-related ancient deltaretroviruses circulated in Asia long before the emergence of BLV. Together, our analyses provide evidence for origin and spatiotemporal dynamics of BLV.
... In remote regions of Australia where Indigenous people were forced to migrate, sepsis tends to stem from a particular cascade of co-infections associated with poverty and crowded living conditions. Human T-cell lymphotropic virus type 1 (HTLV-1) is a leukemia lymphoma causing retrovirus of Asian and African origin that is rare worldwide but endemic in the Reserve System lands in Central Australia (Einsiedel & Fernandes, 2008;Jegado et al., 2019). HTLV-1 predisposes hosts to scabies (Sarcoptes scabiei), threadworm (Strongyloides stercoralis) and pneumonia. ...
Article
COVID‐19 has highlighted a brutal reality known for decades, that Black, Indigenous, and People of Color bear a disproportionate burden of US annual sepsis cases. While plentiful research funds have been spent investigating genetic reasons for racial disparities in sepsis, an abundance of research shows that sepsis incidence and mortality maps to indicators of colonial practices including residential segregation, economic and marginalization sepsis, and denial of care. Here we argue that sepsis risk is an immunological embodiment of racism in colonial states, that the factors contributing to sepsis disparities are insidious and systemic. We show that regardless of causative pathogen, or host ancestry, racialized people get and die of sepsis most frequently in a pattern repeatedly reiterated worldwide. Lastly, we argue that while alleviation of sepsis disparities requires radical, multiscale intervention, biological anthropologists have a responsibility in this crisis. While some of us can harness our expertise to take on the ground action in sepsis prevention, all of us can leverage our positions as the first point of contact for in depth human biology instruction on most college campuses. As a leading cause of death worldwide, and a syndrome that exhibits the interplay between human physiology, race and environment, sepsis is at the nexus of major themes in biological anthropology and is a natural fit for the field's curriculum. In adopting a discussion of race and sepsis in our courses, we not only develop new research areas but increase public awareness of both sepsis and the factors contributing to uneven sepsis burden.
Article
The Human T-cell leukaemia virus type 1 (HTLV-1) is a pathogenic retrovirus that persists as a provirus in the genome of infected cells and can lead to adult T-cell leukaemia (ATL). Worldwide, more than 10 million people are infected and approximately 5% of these individuals will develop ATL, a highly aggressive cancer that is currently incurable. In the last years, genome editing tools have emerged as promising antiviral agents. In this proof-of-concept study, we used substrate linked directed evolution (SLiDE) to engineer Cre-derived site-specific recombinases to excise the HTLV-1 proviral genome from infected cells. We identified a conserved loxP-like sequence (loxHTLV) present in the long terminal repeats of the majority of virus isolates. After 181 cycles of SLiDE, we isolated a designer-recombinase (designated RecHTLV), which efficiently recombines the loxHTLV sequence in bacteria and human cells with high specificity. Expression of RecHTLV in human Jurkat T-cells resulted in antiviral activity when challenged with an HTLV-1 infection. Moreover, expression of RecHTLV in chronically infected SP cells led to the excision of HTLV-1 proviral DNA. Our data suggest that recombinase mediated excision of the HTLV-1 provirus represents a promising approach to reduce proviral load in HTLV-1 infected individuals, potentially preventing the development of HTLV-1 associated diseases.
Article
Full-text available
A small proportion of human T-cell leukemia virus type-1 (HTLV-1)-infected individuals develop adult T-cell leukemia/lymphoma, a chemotherapy-resistant lymphoproliferative disease with a poor prognosis. HTLV-1-specific cytotoxic T lymphocytes (CTLs), potential anti-tumor/virus effectors, are impaired in adult T-cell leukemia/lymphoma patients. Here, using Japanese monkeys naturally infected with simian T-cell leukemia/T-lymphotropic virus type-1 (STLV-1) as a model, we demonstrate that short-term-cultured autologous peripheral blood mononuclear cells (PBMCs) can serve as a therapeutic vaccine to activate such CTLs. In a screening test, STLV-1-specific CTL activity was detectable in 8/10 naturally STLV-1-infected monkeys. We conducted a vaccine study in the remaining two monkeys with impaired CTL responses. The short-term-cultured PBMCs of these monkeys spontaneously expressed viral antigens, in a similar way to PBMCs from human HTLV-1 carriers. The first monkey was subcutaneously inoculated with three-day-cultured and mitomycin C (MMC)-treated autologous PBMCs, and then boosted with MMC-treated autologous STLV-1-infected cell line cells. The second monkey was inoculated with autologous PBMC-vaccine alone twice. In addition, a third monkey that originally showed a weak STLV-1-specific CTL response was inoculated with similar autologous PBMC-vaccines. In all three vaccinated monkeys, marked activation of STLV-1-specific CTLs and a mild reduction in the STLV-1 proviral load were observed. Follow-up analyses on the two monkeys vaccinated with PBMCs alone indicated that STLV-1-specific CTL responses peaked at 3-4 months after vaccination, and then diminished but remained detectable for more than one year. The significant reduction in the proviral load and the control of viral expression were associated with CTL activation but also diminished 6 and 12 months after vaccination, respectively, suggesting the requirement for a booster. The vaccine-induced CTLs in these monkeys recognized epitopes in the STLV-1 Tax and/or Envelope proteins, and efficiently killed autologous STLV-1-infected cells in vitro. These findings indicated that the autologous PBMC-based vaccine could induce functional STLV-1-specific CTLs in vivo.
Preprint
Full-text available
Article
Full-text available
HTLV-1 is an oncovirus causing ATL and other inflammatory diseases such as HAM/TSP and HU in about 5% of infected individuals. It is also known that HTLV-1-infected cells maintain a disease-free, immortalized, latent state throughout the lifetimes of about 95% of infected individuals. We believe that the stable maintenance of disease-free infected cells in the carrier is an intrinsic characteristic of HTLV-1 that has been acquired during its evolution in the human life cycle. We speculate that the pathogenesis of the virus is ruled by the orchestrated functions of viral proteins. In particular, the regulation of Rex, the conductor of viral replication rate, is expected to be closely related to the viral program in the early active viral replication followed by the stable latency in HTLV-1 infected T cells. HTLV-1 and HIV-1 belong to the family Retroviridae and share the same tropism, e.g., human CD4⁺ T cells. These viruses show significant similarities in the viral genomic structure and the molecular mechanism of the replication cycle. However, HTLV-1 and HIV-1 infected T cells show different phenotypes, especially in the level of virion production. We speculate that how the activity of HTLV-1 Rex and its counterpart HIV-1 Rev are regulated may be closely related to the properties of respective infected T cells. In this review, we compare various pathological aspects of HTLV-1 and HIV-1. In particular, we investigated the presence or absence of a virally encoded “regulatory valve” for HTLV-1 Rex or HIV-1 Rev to explore its importance in the regulation of viral particle production in infected T cells. Finally, wereaffirm Rex as the key conductor for viral replication and viral pathogenesis based on our recent study on the novel functional aspects of Rex. Since the activity of Rex is closely related to the viral replication rate, we hypothesize that the “regulatory valve” on the Rex activity may have been selectively evolved to achieve the “scenario” with early viral particle production and the subsequent long, stable deep latency in HTLV-1 infected cells.
Article
Full-text available
Background: Primate T-lymphotropic viruses type 1 (PTLV-1) are complex retroviruses infecting both human (HTLV-1) and simian (STLV-1) hosts. They share common epidemiological, clinical and molecular features. In addition to the canonical gag, pol, env retroviral genes, PTLV-1 purportedly encodes regulatory (i.e. Tax, Rex, and HBZ) and accessory proteins (i.e. P12/8, P13, P30). The latter have been found essential for viral persistence in vivo. Methodology/principal findings: We have isolated a STLV-1 virus from a bonnet macaque (Macaca radiata-Mra18C9), a monkey from India. The complete sequence was obtained and phylogenetic analyses were performed. The Mra18C9 strain is highly divergent from the known PTLV-1 strains. Intriguingly, the Mra18C9 lacks the 3 accessory open reading frames. In order to determine if the absence of accessory proteins is specific to this particular strain, a comprehensive analysis of the complete PTLV-1 genomes available in Genbank was performed and found that the lack of one or many accessory ORF is common among PTLV-1. Conclusion: This study raises many questions regarding the actual nature, role and importance of accessory proteins in the PTLV-1 biology.
Article
Full-text available
Simian T-Leukemia Virus type 1 and Simian Foamy Virus infect non-human primates. While STLV-1, as HTLV-1, causes Adult T-cell Leukemia/lymphoma, SFV infection is asymptomatic. Both retroviruses can be transmitted from NHPs to humans through bites that allow contact between infected saliva and recipient blood. Because both viruses infect CD4+ T-cells, they might interfere with each other replication, and this might impact viral transmission. Impact of STLV-1 co-infection on SFV replication was analyzed in 18 SFV-positive/STLV-1-negative and 18 naturally SFV/STLV-1 co-infected Papio anubis. Even if 9 animals were found STLV-1-positive in saliva, STLV-1 PVL was much higher in the blood. SFV proviruses were detected in the saliva of all animals. Interestingly, SFV proviral load was much higher in the blood of STLV-1/SFV co-infected animals, compared to STLV-1-negative animals. Given that soluble Tax protein can enter uninfected cells, we tested its effect on foamy virus promoter and we show that Tax protein can transactivate the foamy LTR. This demonstrates that true STLV-1 co-infection or Tax only has an impact on SFV replication and may influence the ability of the virus to be zoonotically transmitted as well as its ability to promote hematological abnormalities.
Article
Full-text available
Going back to their discovery in the early 1980s, both the Human T-cell Leukemia virus type-1 (HTLV-1) and the Human Immunodeficiency Virus type-1 (HIV-1) greatly fascinated the virology scene, not only because they were the first human retroviruses discovered, but also because they were associated with fatal diseases in the human population. In almost four decades of scientific research, both viruses have had different fates, HTLV-1 being often upstaged by HIV-1. However, although being very close in terms of genome organization, cellular tropism, and viral replication, HIV-1 and HTLV-1 are not completely commutable in terms of treatment, especially because of the opposite fate of the cells they infect: death versus immortalization, respectively. Nowadays, the antiretroviral therapies developed to treat HIV-1 infected individuals and to limit HIV-1 spread among the human population have a poor or no effect on HTLV-1 infected individuals, and thus, do not prevent the development of HTLV-1-associated diseases, which still lack highly efficient treatments. The present review mainly focuses on the course of HTLV-1 infection, from the initial infection of the host to diseases development and associated treatments, but also investigates HIV-1/HTLV-1 co-infection events and their impact on diseases development.
Article
Full-text available
Human T-cell leukemia virus type 1 (HTLV-1) infects mainly CD4⁺CCR4⁺ effector/memory T cells in vivo. However, it remains unknown whether HTLV-1 preferentially infects these T cells or this virus converts infected precursor cells to specialized T cells. Expression of viral genes in vivo is critical to study viral replication and proliferation of infected cells. Therefore, we first analyzed viral gene expression in non-human primates naturally infected with simian T-cell leukemia virus type 1 (STLV-1), whose virological attributes closely resemble those of HTLV-1. Although the tax transcript was detected only in certain tissues, Tax expression was much higher in the bone marrow, indicating the possibility of de novo infection. Furthermore, Tax expression of non-T cells was suspected in bone marrow. These data suggest that HTLV-1 infects hematopoietic cells in the bone marrow. To explore the possibility that HTLV-1 infects hematopoietic stem cells (HSCs), we analyzed integration sites of HTLV-1 provirus in various lineages of hematopoietic cells in patients with HTLV-1 associated myelopathy/tropical spastic paraparesis (HAM/TSP) and a HTLV-1 carrier using the high-throughput sequencing method. Identical integration sites were detected in neutrophils, monocytes, B cells, CD8⁺ T cells and CD4⁺ T cells, indicating that HTLV-1 infects HSCs in vivo. We also detected Tax protein in myeloperoxidase positive neutrophils. Furthermore, dendritic cells differentiated from HTLV-1 infected monocytes caused de novo infection to T cells, indicating that infected monocytes are implicated in viral spreading in vivo. Certain integration sites were re-detected in neutrophils from HAM/TSP patients at different time points, indicating that infected HSCs persist and differentiate in vivo. This study demonstrates that HTLV-1 infects HSCs, and infected stem cells differentiate into diverse cell lineages. These data indicate that infection of HSCs can contribute to the persistence and spread of HTLV-1 in vivo.
Article
Full-text available
There are four lineages of primate T-cell lymphocytic viruses (human T-cell lymphocytic virus [HTLV]/simian T-cell lymphocytic virus [STLV]), which are further divided into subtypes. To date, there is only one full-length HTLV-1 subtype b genome available. Here, we report the genome of a new STLV-1 subtype b from a 43-year-old male gorilla with T-cell lymphoma.
Article
Full-text available
Baboons naturally infected with simian T lymphotropic virus (STLV) are a potentially useful model system for the study of vaccination against human T lymphotropic virus (HTLV). Here we expanded the number of available full-length baboon STLV-1 sequences from one to three and related the T cell responses that recognize the immunodominant Tax protein to the tax sequences present in two individual baboons. Continuously growing T cell lines were established from two baboons, animals 12141 and 12752. Next-generation sequencing (NGS) of complete STLV genome sequences from these T cell lines revealed them to be closely related but distinct from each other and from the baboon STLV-1 sequence in the NCBI sequence database. Overlapping peptides corresponding to each unique Tax sequence and to the reference baboon Tax sequence were used to analyze recognition by T cells from each baboon using intracellular cytokine staining (ICS). Individual baboons expressed more gamma interferon and tumor necrosis factor alpha in response to Tax peptides corresponding to their own STLV-1 sequence than in response to Tax peptides corresponding to the reference baboon STLV-1 sequence. Thus, our analyses revealed distinct but closely related STLV-1 genome sequences in two baboons, extremely low heterogeneity of STLV sequences within each baboon, no evidence for superinfection within each baboon, and a ready ability of T cells in each baboon to recognize circulating Tax sequences. While amino acid substitutions that result in escape from CD8⁺ T cell recognition were not observed, premature stop codons were observed in 7% and 56% of tax sequences from peripheral blood mononuclear cells from animals 12141 and 12752, respectively. IMPORTANCE It has been estimated that approximately 100,000 people suffer serious morbidity and 10,000 people die each year from the consequences associated with human T lymphotropic virus (HTLV) infection. There are no antiviral drugs and no preventive vaccine. A preventive vaccine would significantly impact the global burden associated with HTLV infections. Here we provide fundamental information on the simian T lymphotropic virus (STLV) naturally transmitted in a colony of captive baboons. The limited viral sequence heterogeneity in individual baboons, the identity of the viral gene product that is the major target of cellular immune responses, the persistence of viral amino acid sequences that are the major targets of cellular immune responses, and the emergence in vivo of truncated variants in the major target of cellular immune responses all parallel what are seen with HTLV infection of humans. These results justify the use of STLV-infected baboons as a model system for vaccine development efforts.
Article
Full-text available
Simian T-lymphotropic virus 1 (STLV-1) enters human populations through contact with nonhuman primate (NHP) bushmeat. We tested whether differences in the extent of contact with STLV-1-infected NHP bushmeat foster regional differences in prevalence of human T-lymphotropic virus 1 (HTLV-1). Using serological and PCR assays, we screened humans and NHPs at two Sub-Saharan African sites where subsistence hunting was expected to be less (Taï region, Côte d'Ivoire [CIV]) or more (Bandundu region, Democratic Republic of the Congo [DRC]) developed. Only 0.7% of human participants were infected with HTLV-1 in CIV (n = 574), and 1.3% of humans were infected in DRC (n = 302). Two of the Ivorian human virus sequences were closely related to simian counterparts, indicating ongoing zoonotic transmission. Multivariate analysis of human demographic parameters and behavior confirmed that participants from CIV were less often exposed to NHPs than participants from DRC through direct contact, e.g., butchering. At the same time, numbers of STLV-1-infected NHPs were higher in CIV (39%; n = 111) than in DRC (23%; n = 39). We conclude that similar ultimate risks of zoonotic STLV-1 transmission-defined as the product of prevalence in local NHP and human rates of contact to fresh NHP carcasses-contribute to the observed comparable rates of HTLV-1 infection in humans in CIV and DRC. We found that young adult men and mature women are most likely exposed to NHPs at both sites. In view of the continued difficulties in controlling zoonotic disease outbreaks, the identification of such groups at high risk of NHP exposure may guide future prevention efforts.
Article
Full-text available
Background Virus transmission from various wild and domestic animals contributes to an increased risk of emerging infectious diseases in human populations. HTLV-1 is a human retrovirus associated with acute T-cell leukemia and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). HTLV-1 originated from ancient zoonotic transmission from nonhuman primates, although cases of zoonotic infections continue to occur. Similar to HTLV-1, the simian counterpart, STLV-1, causes chronic infection and leukemia and lymphoma in naturally infected monkeys, and combined are called primate T-lymphotropic viruses (PTLV-1). However, other clinical syndromes typically seen in humans such as a chronic progressive myelopathy have not been observed in nonhuman primates. Little is known about the development of neurologic and inflammatory diseases in human populations infected with STLV-1-like viruses following nonhuman primate exposure. ResultsWe performed detailed laboratory analyses on an HTLV-1 seropositive patient with typical HAM/TSP who was born in Liberia and now resides in the United States. Using a novel droplet digital PCR for the detection of the HTLV-1 tax gene, the proviral load in PBMC and cerebrospinal fluid cells was 12.98 and 51.68 %, respectively; however, we observed a distinct difference in fluorescence amplitude of the positive droplet population suggesting possible mutations in proviral DNA. A complete PTLV-1 proviral genome was amplified from the patient’s PBMC DNA using an overlapping PCR strategy. Phylogenetic analysis of the envelope and LTR sequences showed the virus was highly related to PTLV-1 from sooty mangabey monkeys (smm) and humans exposed via nonhuman primates in West Africa. Conclusions These results demonstrate the patient is infected with a simian variant of PTLV-1, suggesting for the first time that PTLV-1smm infection in humans may be associated with a chronic progressive neurologic disease.
Article
HTLV-1 causes Adult T cell Leukemia/Lymphoma (ATLL) in humans. We describe an ATL-like disease in a 9 year-old female baboon naturally infected with STLV-1 (the simian counterpart of HTLV-1), with a lymphocyte count over 10¹⁰/L, lymphocytes with abnormal nuclear morphology, and pulmonary and skin lesions. The animal was treated with a combination of AZT and alpha interferon. Proviral load (PVL) was measured every week. Because the disease continued to progress, the animal was euthanized. Abnormal infiltrates of CD3⁺CD25⁺ lymphocytes and Tax-positive cells were found by histological analyses in both lymphoid and non-lymphoid organs. PVL was measured and clonal diversity was assessed by LM-PCR (Ligation-Mediated Polymerase Chain Reaction) and high throughput sequencing, in blood during treatment and in 14 different organs. The highest PVL was found in lymph nodes, spleen and lungs. One major clone and a number of intermediate abundance clones were present in blood throughout the course of treatment, and in organs. These results represent the first multi-organ clonality study in ATLL. We demonstrate a previously undescribed clonal complexity in ATLL. Our data reinforce the usefulness of natural STLV-1 infection as a model of ATLL.