You are on page 1of 12

REVIEW

Eckstrand et al., Journal of General Virology 2017;98:1985–1996


DOI 10.1099/jgv.0.000866

Central and peripheral reservoirs of feline immunodeficiency


virus in cats: a review
Chrissy D. Eckstrand,1,* Ellen E. Sparger2 and Brian G. Murphy3

Abstract
Infection with feline immunodeficiency virus (FIV), a lentivirus similar to human immunodeficiency virus (HIV), results in
lifelong viral persistence and progressive immunopathology in the cat. FIV has the ability to infect and produce infectious
virus in a number of different cell types. FIV provirus can also be maintained in a replication-competent but transcriptionally
quiescent state, facilitating viral persistence over time. Immediately after the initial infection, FIV infection quickly
disseminates to many anatomical compartments within the host including lymphoid organs, gastrointestinal tract and brain.
Collectively, the anatomic and cellular compartments that harbour FIV provirus constitute the viral reservoir and contain foci
of both ongoing viral replication and transcriptionally restricted virus that may persist over time. The relative importance of
the different phenotypes observed for infected cells, anatomic compartment, replication status and size of the reservoir
represent crucial areas of investigation for developing effective viral suppression and eradication therapies. In this review,
we discuss what is currently known about FIV reservoirs, and emphasize the utility of the FIV-infected cat as a model for the
HIV-infected human.

INTRODUCTION cohabitation with other cats and concurrent disease status


have been proposed [7–12]. Efforts to control lentiviral infec-
Feline immunodeficiency virus (FIV) is a lentivirus that
tions primarily focus on two broad strategies: preventing new
shares similarities to human immunodeficiency virus (HIV)
infections and suppressing viral replication in already infected
and is present in both domestic and feral cat (Felis catus)
individuals, both of which are particularly relevant for HIV-
populations worldwide. FIV prevalence varies by geographic
infected humans. The complete elimination of virus-infected
region with the incidence in North America reported at
cells from the host is a laudable goal. However, practical and
2.5 % (2006), 5.4 % in Bangkok, Thailand (2014), 9.5 % in
effective viral eradication strategies do not yet exist for animals
Ankara, Turkey (2013), 23.2 % in Japan (2008) and 31.1 %
or humans infected with lentiviruses in spite of highly effective
in peninsular Malaysia (2012), equating to millions of FIV-
viral suppression therapies and several decades of research
infected animals worldwide [1–5].
focused on the pathogenesis of lentiviral agents [13]. The
Infection with FIV results in lifelong viral persistence and pro- identification of permissive cell types and anatomical com-
gressive immune dysfunction; however the clinical outcomes partments harbouring virus have been important goals in
of FIV-infected cats are variable, with many FIV-infected cats research related to immunodeficiency-inducing lentiviruses.
living approximately normal lifespans [6]. Although FIV has Such information is critical to the development of effective
been associated with unfavourable outcomes such as opportu- eradication therapies which depend upon the tissue penetra-
nistic infections, neoplasia, wasting and death, the precise tion of various pharmaceutical agents. The relative importance
determinants of morbidity remain unclear. Contributory fac- of the specific cell types and anatomical compartments that
tors including viral/host genetics, age of host, viral load constitute the lentiviral reservoir in an infected host remain
during acute virus infection, housing environment, stress, controversial [13, 14].

Received 28 March 2017; Accepted 15 June 2017


Author affiliations: 1Veterinary Microbiology and Pathology, College of Veterinary Medicine, 4003 Animal Disease Biotechnology Facility, Washington
State University, Pullman, WA 99163, USA; 2Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, 3115
Tupper Hall, Davis, CA 95616, USA; 3Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, 4206 Vet Med 3A,
University of California, Davis, CA 95616, USA.
*Correspondence: Chrissy D. Eckstrand, ceckstrand@vetmed.wsu.edu
Keywords: FIV; latency; lentivirus; reservoir; tissue; viral persistence.
Abbreviations: FIV, feline immunodeficiency virus; HIV, human immunodeficiency virus; SIV, simian immunodeficiency virus; FAIDS, feline acquired
immunodeficiency syndrome; Tfh, T follicular helper; IF, immunofluorescence; IHC, immunohistochemistry; RT, reverse-transcriptase; CNS, central
nervous system; ART, antiretroviral therapy; GALT, gut-associated lymphoid tissue; PBMCs, peripheral blood mononuclear cells; SPF, specific patho-
gen free; qPCR, quantitative polymerase chain reaction; VOA, viral outgrowth assay; SAHA, suberoylanilide hydroxamic acid; VPA, valproic acid; TSA,
trichostatin; NaB, sodium butyrate; DZNep, 3-deazanoplanocin A; PKC, protein kinase-C; MLN, mesenteric lymph node.

000866

1985
Eckstrand et al., Journal of General Virology 2017;98:1985–1996

Multiple factors play roles in defining a cell’s permissiveness FIV RECEPTOR USAGE
to lentiviral infection and productive replication and include Early reports revealed the presence of FIV proviral DNA in
the appropriate membrane receptors for virus attachment, specific cell subsets including CD4+ and CD8+ T cells, mac-
fusion and entry, a temporally appropriate milieu of nuclear rophages and dendritic cells by sorting of these cell popula-
transcription factors, activation state, target cell subset, tissue tions from experimentally infected cats or analysis of tissues
micro-environment, and presence of specific host cellular by immunohistochemistry (IHC) [26–31]. Subsequent stud-
restriction factors like tetherin, APOBEC3 family and others ies revealed that FIV utilizes two cell surface receptors iden-
[11, 13, 15, 16]. These same factors also determine the identity tified as the costimulatory molecule CD134 (OX40) and
of cells that serve as reservoirs for persistent latent virus [13]. the chemokine receptor CXCR4 for cellular attachment and
This review discusses what is currently known about the cellu- entry into these specific cellular targets [32–34]. Primary
lar and anatomical reservoirs in the FIV-infected cat and point binding of the cell by the virion is mediated between the
out similarities shared by the FIV- and HIV-infected host. For FIV Env-SU protein and the cell surface receptor CD134, a
the purpose of this review, reservoirs relevant to viral patho- costimulatory marker expressed by activated CD4 T cells.
genesis will be defined as cells and tissues that persistently har- Binding instigates a conformational change in Env, which
bour replication-competent provirus. The FIV-infected cat facilitates subsequent binding between the cell surface
offers the opportunity to investigate the virologic and cellular expressed chemokine receptor CXCR4 and the V3 region of
features of central viral reservoirs that are typically inaccessible Env-SU [35, 36]. Binding of a primary cell surface receptor
in living HIV-infected humans, and may also serve as a model to facilitate Env interactions with a surface chemokine
for assessing the effectiveness of viral suppression and eradica- receptor is a mode of cell entry that is also well character-
tion therapies. ized for other immunodeficiency-inducing lentiviruses such
as HIV-1 and SIV [37, 38].
FIV BACKGROUND CD134 is a transmembrane glycoprotein within the tumour
Feline immunodeficiency virus was initially isolated in the necrosis factor superfamily, and serves as the initial binding
mid-1980s from a cohort of group-housed domestic cats in receptor equivalent to the CD4 cell surface receptor in HIV
northern California exhibiting a variety of non-specific ill- binding [35]. Human and mouse CD134 are predominantly
nesses such as gingivitis, enteritis, dermatitis, weight loss and expressed on activated CD4+ T cells including regulatory T
death [17]. Virus isolation, electron microscopy, reverse tran- cells and T follicular helper (Tfh) cells, and more transiently
scriptase enzyme assays and Western blot analyses identified a expressed on activated CD8+ T cells. CD134 expression by
T-lymphotropic retrovirus in the cats with clinical disease [17, natural killer (NK) cells, NKT cells and neutrophils has also
18]. Subsequent to this discovery, investigations into naturally been reported [39–41]. Currently, expression of feline
and experimentally infected domestic cats have revealed a CD134 has been demonstrated on the surface of CD4+ T
plethora of information about the virus and disease pathogen- and CD8+ T cells, and B220 (CD45R)+ B cells, but not on
esis; as a result, the FIV-infected cat has become a well- peripheral blood-derived monocytes [42, 43]. However,
recognized translational model for investigation of HIV path- feline monocyte-derived macrophages, tissue-derived CD14
ogenesis, as well as development of antiviral therapeutics and + macrophages and activated CD8+ T cells were shown to
vaccines. The FIV-infected cat is the only naturally occurring express CD134 surface expression that was further enhanced
HIV animal model for a clinical immunodeficiency that is with lipopolysaccharide activation [33, 42]. In a separate
characterized by a progressive CD4+ T cell depletion, oppor- study, cellular CD134 mRNA was detected in feline T and B
tunistic infections and various entities including wasting and lymphocytes, dendritic cells and macrophages; the highest
neurological disorders [19, 20]. As such, FIV is distinguished expression, which increased with activation, was detected in
from other experimental animal models such as simian immu- activated T lymphocytes [43]. Overall, CD134 expression
nodeficiency virus (SIV)-infection of non-natural hosts correlated with immune cell populations shown to be sus-
including the Asian macaque species and the HIV-infected ceptible to FIV infection in vitro or to be positive for viral
humanized mouse [21]. Similar to HIV-infected humans, FIV DNA in the infected cat.
infection follows a relatively predictable course of disease Early reports showed that different CD134 binding-domain
through three phases: the acute, chronic asymptomatic and patterns may be viral strain-dependent [35, 44, 45]. Further-
terminal feline acquired immunodeficiency syndrome stage more, CD134 domains required for FIV Env binding and
(FAIDS) [22–24]. Numerous reports have described infection with more pathogenic FIV strains were mapped to
the virologic and immunopathologic aspects of acute and early two cysteine-rich domains (CRD1 and CRD2), whereas less
asymptomatic phases of FIV infection through experimental pathogenic strains only required binding to CRD1 based on
inoculation studies. However, investigation of the late asymp- in vitro analyses. These findings agreed with evidence for
tomatic phase and transition into FAIDS has been primarily FIV Env-CD134 binding patterns changing over time in
restricted to clinical cohorts of cats naturally infected with FIV individually infected animals, which may ultimately influ-
[7, 23, 25]. This is perhaps due to the cost of maintaining and ence viral cell surface affinity due to nucleotide base changes
housing experimentally infected cats in specific pathogen free in env and reflect isolates sensitive to circulating anti-
(SPF) facilities for prolonged time frames. CD134 autoantibodies [44–46]. Recent studies further

1986
Eckstrand et al., Journal of General Virology 2017;98:1985–1996

characterized FIV Env changes associated with CD134 Accordingly, HIV-1 and FIV utilize similar strategies for
binding of distinctive FIV isolates that emerge in naturally entry of target cells by Env binding of a primary cell surface
infected cats during disease progression [47]. Variants char- receptor, CD4 and CD134, respectively, that facilitates Env
acterized as Env recombinant viruses have also been interactions with either the CCR5 (HIV-1 only) or CXCR4
described [48]. Notably, specific changes in the env gene (HIV-1 and FIV) chemokine coreceptors [37, 49]. Further-
sequence mapped over longitudinal time points of infection more, targeting CD4+ T cells through either CD4 or CD134
resulted in FIV variants that lost dependence for binding to surface proteins results in a similar immunopathogenesis,
the CRD2 domain of CD134 for virus entry. These CRD2- characterized by a progressive CD4+ T cell depletion, lym-
independent isolates, which are characteristic of the later phoid depletion and immune exhaustion during the chronic
stages of FIV infection, were also shown to be less efficient phases of HIV-1 and FIV infections, respectively.
for transmission and replication during acute infection
when compared to CRD2-dependent isolates in experimen- CELL RESERVOIRS OF FIV
tal infection studies. These findings generated a hypothesis
Specific cell reservoirs of FIV are discussed with the under-
that CRD2-dependent FIV isolates, which are efficiently
standing that a cellular reservoir contains integrated replica-
transmitted in vivo and able to achieve higher virus
tion-competent provirus and persists over time. The biology
loads during early infection, may be analogous to CCR5-
of FIV cellular reservoirs is complex and dynamic. Critical
dependent HIV-1 isolates that also transmit much more
factors include different cell types that are permissive to FIV
efficiently during primary HIV-1 infection of humans when
infection, infecting strain of FIV, changes in absolute CD4+
compared to later-emerging CXCR4-tropic variants in HIV- and CD8+ T cell numbers, cell surface receptor expression
1 infection [49, 50]. Specific CD4+ T cell subsets that may modulation over time, stage of infection, evolution of the
be specific targets for these CRD2-dependent FIV isolates viral genotype over time in the infected host, host-adaptive
and their role in viral pathogenesis remain to be immune responses and mechanisms of viral latency. Similar
determined. to HIV-1, FIV infects and rapidly expands during the acute
Flow cytometric analysis revealed that chemokine receptor stage of the disease, and then persists in peripheral blood
CXCR4 is a co-receptor shared by both FIV and HIV-1 and and tissue CD4+ T cells [26, 27, 52, 57]. However, CD4+ T
is expressed on feline peripheral blood-derived monocytes, cells are not the only relevant reservoirs as the virus also
B-cells and activated T cells [51, 52]. These findings for infects and disseminates to a wide variety of other leuko-
feline CXCR4+ cell subsets overall coincide with cells that cytes during the acute stage of infection, including CD8+ T
express FIV primary receptor feline CD134, and also cells, B-cells, monocytes/macrophages and other long-lived
account for most cellular subsets shown to infected with cells such as dendritic cells and megakaryocytes [26, 27, 29,
FIV in vivo, as described below. FIV Env domains responsi- 31, 58, 59]. Evidence supporting that these leukocytes are
ble for binding of CXCR4 and CD134 have also been susceptible and permissive to FIV include PCR for proviral
mapped and found to be independent of each other, as DNA and viral RNA, in situ hybridization for intracellular
expected [36, 53]. Additionally, CXCR4 expression has been FIV genomic RNA and/or DNA, immunofluorescence (IF)
detected in lymph node, thymic and bone marrow-derived or IHC assessment of tissues for detection of viral antigen,
CD4+, CD8+ T lymphocytes, CD21+ B lymphocytes and and either reverse-transcriptase (RT) activity assays
unfractionated CD4-/CD8-/CD21- cells [54]. Results from or enzyme-linked immunosorbant assay (ELISA) for detec-
this report also revealed FIV infection in both CXCR4+ and tion of extracellular viral protein. Table 1 summarizes key
CXCR4- cell populations, as determined by quantitative studies that have identified FIV cell reservoirs and the
polymerase chain reaction (qPCR) assay of isolated cell pop- modalities used to detect infection or susceptibility to infec-
ulations for FIV proviral DNA. These results suggested the tion. It is important to note that the leukocyte subsets that
possibility that FIV may be capable of infecting cells in a have proven most consistently infected in vivo and suscepti-
CXCR4-independent manner, although this finding has not ble to infection in vitro are CD4+ T cells and monocyte/
been tested further or confirmed by subsequent studies. macrophages, which are also target cell reservoirs for HIV
Interestingly, laboratory-adapted FIV isolates have been [13, 49, 54, 60–62]. Evidence thus far is conflicting regard-
shown to be capable of infecting cells independent of ing productive infection of dendritic cells in vivo [29, 30, 54,
CD134 by use of heparin sulphate proteoglycans (HSPGs) 63], although multiple studies support productive FIV infec-
and DC-SIGN as binding receptors to promote infection tion of cultivated dendritic cells [43, 63, 64]. Likewise,
and facilitate binding of CXCR4 in the absence of CD134 reports may demonstrate productive HIV-1 infection of
dendritic cells in vitro [65], but in vivo analyses suggest that
[53]. FIV Env binding of HSPG and DC-SIGN are proper-
extracellular virions bound to the cell surface of dendritic
ties shared with HIV-1 [55, 56] wherein HIV-1 Env binding
cells are the primary mode by which these cells transmit
of these receptors on dendritic cells may facilitate transmis-
virus infection to CD4 T cells [13, 66]. Evidence for produc-
sion of HIV-1 virions to T cell targets.
tive replication of other subsets such as CD8+ T cells and B
However, usage of these alternative receptors appears to be cells in vivo or in vitro has not been reported. These findings
restricted to laboratory-adapted FIV isolates and the role of point once more to the similarities in cell tropism shared by
these receptors in FIV pathogenesis is currently unknown. FIV and HIV-1.

1987
Table 1. Summary of scientific reports that identify cellular and tissue reservoirs of feline immunodeficiency virus
Author and Year Infection period FIV-infected cell type(s) identified FIV-infected tissue(s) identified Method(s) of evaluation FIV strain
(derived compartment, infection type)

Pedersen et al. [17] Unknown T-lymphocyte (blood, in vivo infection) NA RT activity, Western Blot, Electron Petaluma
microscopy
Brown et al. [59] NA CD4, CD8 T lymphocytes NA RT activity (supe) Petaluma, Peeper
(blood, in vitro infection) IFA (FIV polyclonal serum)
Dow et al. [67] NA Astrocytes, microglia NA IFA (FIV polyclonal serum) p26 ELISA (supe) Petaluma
(primary cell culture, in vitro infection)
Beebe et al. [103] 119–335 days Megakaryocytes, mononuclear cells Bone marrow ISH (proviral and viral RNA gag, pol, env) Petaluma
(tissue, in vivo infection)
English et al. [26] 2–4 weeks and CD4, CD8, Ig+ (blood, in vivo infection) NA PCR (proviral gag) FIV-NCSU1, Petaluma, Mt. Airy
12–18 months RT activity (supe)
Toyosaki et al. [99] 5 months CD4, FDC, CD8 (tissue, in vivo infection) Lymph node IHC (gag polyclonal ab) TM-1, KYO-1, Petaluma
Dua et al. [81] 1–12 weeks NA CNS, tonsil, thymus, bone marrow, lymph PCR (proviral gag) Petaluma
nodes, liver, spleen, kidney, intestine, lung,
salivary gland
Beebe et al. [31] 3 months CD3, macrophages (tissue, in vivo infection) Bone marrow, thymus, lymph nodes, tonsil, ISH (proviral and viral RNA gag, pol, env), Petaluma
spleen, intestine, kidney, lung, liver IHC (CD3, Mac387)
Park et al. [97] 2–6 weeks Epithelial cells of interlobular ducts (tissue, in Salivary gland IHC (p26), PCR (proviral env) West Aust T91
vivo infection)
Dean et al. [27] 2–306 weeks CD4, CD8, CD21 (blood and tissue, in vivo Lymph node PCR (proviral, viral RNA gag), p24 ELISA Petaluma
infection) (supe)
Burkhard et al. [95] 8 weeks PBMCs, LNC, IEL (tissue, in vivo infection) Thymus, lymph node, spleen, intestine, Virus isolation, PCR (proviral, viral RNA gag), FIV-B-2542
vaginal mucosa, rectal mucosa ISH (whole genome)
Woo et al. [57] 2–20 weeks CD1, CD21/CD22, CD4/CD8 (thymus, in NA PCR (proviral and viral RNA gag) Petaluma
vivo infection)
Rogers and Hoover [94] 5–9 weeks (foetus) NA PBMCs, brain, thymus, bone marrow, MLN, PCR (proviral gag) FIV-B-2542
spleen, liver
Dow et al. [58] 12–16 weeks Monocytes (blood, in vivo infection) NA ICC (FIV polyclonal serum), IF (FIV p26) FIV-B (2542, 2560, 2561)
PCR (FIV proviral env), ELISA (supe and FIV-AB (2531,2546)

1988
lysate FIV p26) FIV-A-Petaluma
Tanabe and Yamamoto [102] 24 weeks–1 year Macrophages and fibroblasts (bone marrow, NA RT (supe) FIV-UK8, Bang, Shi, Petaluma
in vivo infection)
Rogers et al. [29] 3 weeks CD3 T cells, macrophages, dendritic cells Lymph node, thymus, spleen, intestine, bone IHC, IFA (FIV polyclonal plasma) FIV-B-2542
(tissue) marrow, liver ISH (gag, envA, envB) FIV Petaluma
Joshi et al. [52] 5 years CD4+CD25+, CD4+CD25- (blood and lymph NA PCR (LTR, LTR-gag, env-gag CJs) p24 Ag NCSU1
node, in vivo and in vitro infections) capture
Hein et al. [107] 14–183 days Microglia (brain, in vivo infection) NA PCR (viral RNA gag) Wo, Womic
Troth et al. [54] 3–4 weeks CD4/CD8, B lymphocyte (CA1.2D6), CXCR NA PCR (proviral gag) FIV-C-Pgmr
+/- (blood, bone marrow, lymph node,
thymus, in vivo infection)
Reggeti et al. [43] NA CD4, CD8, CD21 (blood, in vitro infection) NA PCR (proviral LTR-gag) p24 ELISA (supe) USgaB01
Eckstrand et al., Journal of General Virology 2017;98:1985–1996

Macrophages, dendritic cells (bone marrow,


in vitro infection)
Sprague et al. [64] NA Dendritic cells (blood, in vitro infection) NA PCR (proviral gag), p26 ELISA, Electron FIV-C-Pgmr
microscopy
Miller et al. [104] 350 days NA Brain PCR (viral RNA gag) Pcenv, PPR, C36
Hood et al. [63] 1–3 years Monocytes, dendritic cells, CD8 lymphocytes NA PCR (undescribed viral RNA in supe) NCSU1
(blood, in vivo infection)
Eckstrand et al. [109] 5.1 years CD4, CD21 (blood and lymph node, in vivo Lymph node PCR (proviral and viral RNA gag), Western FIV-C-Pgmr
infection) blot, IHC (polyclonal FIV plasma), ex vivo
reactivation
Eckstrand et al. [92] 6.1 years CD4, CD8, CD21 (blood, lymph node, spleen, Lymph node, spleen, small intestine PCR (proviral and viral RNA gag), ISH (whole FIV-C-Pgmr
in vivo infection) genome), ex vivo reactivation

ab, antibody; ELISA, enzyme-linked immunosorbent assay; ICC, immunocytochemistry; IFA, immunofluorescent antibody; IHC, immunohistochemistry; ISH, in situ hybridization; NA, not
applicable; PCR, polymerase chain reaction; PBMC, peripheral blood mononuclear cell; RT, reverse transcriptase; supe, supernatant.
Eckstrand et al., Journal of General Virology 2017;98:1985–1996

FIV is capable of infecting and replicating in cell types other CD4+ T cell counts continue to progressively decline to
than traditional leukocytes. Feline central nervous system very low levels in blood and lymphoid tissues and may be
(CNS)-derived glial cells, including astrocytes and micro- associated with declining peripheral CD8+ T cell frequen-
glia, have been shown to support FIV replication in vivo cies and clinical signs of immunodeficiencies [7, 19, 83].
and in vitro [67–71]. Infection of the feline CNS by FIV is Interestingly, significant declines in CD4+ T cells counts are
proposed to result from trafficking of infected B and T not always associated with clinical signs of acquired immu-
cells, as well as macrophages, through the blood–brain bar- nodeficiencies and other factors such as environmental
rier and into the choroid plexus based on experimental cat stresses may determine disease onset [7, 24, 79].
inoculation studies [72] and ex vivo choroid plexus epithe-
lial cultures [73]. It is perhaps not surprising that microglia Early studies investigating FIV-infected target cell popula-
can support FIV infection, as these cells arise from bone tions during experimental infection relied on IHC analysis
marrow myeloid progenitor cells similar to other tissue- with a limited array of cell surface markers, in situ hybrid-
based macrophages. However, it is puzzling that astrocytes ization for viral nucleic markers and PCR analysis fre-
seem to support FIV replication, as these glial cells are quently dependent on nested-PCR protocols. Based on
derived from neuroectoderm. Billaud et al. demonstrated these reports [26, 27, 31, 81], highest viral loads were
that astrocytes not only support FIV replication in vitro, but detected in CD4+ T cells. However, findings regarding shifts
that viral replication is dependent upon the env sequence in proviral DNA loads to other subsets varied between these
and that the replication rate in astrocytes differs between early studies to reflect greater loads in B cells, CD8+ T cells
FIV strains [70]. However, other studies failed to show pro- or macrophages depending on the virus isolate tested. Later
ductive FIV infection of astrocytes [74]. Similarly, findings reports and summaries [29, 54, 60, 79, 84, 85] also show
for HIV-1 infection of astrocytes have been conflicting. variable findings. Yet, overall, these later studies reported
Astrocytes were shown to support HIV infection in both in that CD4+ T cells followed by CD21+ B cells and mono-
vivo and in vitro studies, with the human mannose receptor cyte/macrophages have the highest proviral DNA loads in
reported to be essential for CD4-independent infection of blood and lymphoid tissue, including the mucosa of the gas-
this cell type [75, 76]. However, more recent studies trointestinal tract. Dendritic cells have also been reported to
reported inconsistent findings regarding HIV infection of be positive for proviral DNA in the few studies that analysed
astrocytes and suggest that infection of astrocytes is highly this subset, although the relevance of FIV infection of den-
restricted, may involve CD81 vesicles and may internalize dritic cells in vivo is still poorly understood [29, 30, 54]. Tar-
virus particles without productive virus replication [77, 78]. geting of CD4+ T cells for a progressive depletion in the
Regardless, the overall findings indicate that astrocyte infec- periphery and in lymphoid tissues along with targeting of
tion or internalization lead to cellular dysfunction and facili- other immune cell subsets including monocyte/macro-
tate transfer and dissemination of HIV-1 within the CNS. phages, and dendritic cells parallels findings reported for
The role and nature of FIV interactions with astrocytes in HIV-1 infection in humans and correlates with the onset of
virus-induced disease of the CNS, and similarities shared a clinical immunodeficiency in the FIV-infected cat.
with HIV, remain to be determined.
There are relatively few reports describing longitudinal FIV TISSUE RESERVOIRS
studies designed to investigate changes within infected leu-
kocyte subsets according to the stage of FIV infection. Evi- From a broader perspective, anatomical sites that harbour
dence that proviral DNA loads may shift between cellular FIV-infected cells have also been an active area of investiga-
subsets as disease progresses, especially as CD4+ T cells are tion in research focused on lentiviral pathogenesis. The
depleted, has been presented in a limited number of reports topic of anatomical reservoirs of lentiviral persistence
[26, 27, 79]. The hallmark of FIV infection in the acute and became particularly important with the advent of antiretro-
early asymptomatic phases of infection is an inverted viral drug therapy (ART) for HIV-infected humans. While
peripheral blood CD4:CD8 T cell ratio initially due to a ART is highly effective for suppressing HIV viral replica-
decrease in CD4+ T cells and in some cases a concurrent tion, there is evidence that incomplete anatomically local-
expansion of CD8+ T cells [60, 80–82]. The abrupt decline ized suppression of viral replication occurs due to
in peripheral CD4+ T cell count typically rebounds, inadequate tissue drug concentrations in particular ana-
although a return to pre-infection values may not be tomic sites such as the brain [86–90]. As is true for cellular
observed. This decline in peripheral CD4+ T cells counts reservoirs, technical challenges impede the identification of
during acute infection has been reported to coincide with tissue reservoirs infected with replication-competent provi-
even more severe declines in CD4+ T cell frequencies in rus. Multiple methodologies have been utilized to identify
lymphoid tissues with the severity dependent on the FIV FIV-infected cells in tissues, including PCR to identify pro-
isolate used for inoculation and route of infection [60]. virus in tissue homogenates, tissue localization of virus
Importantly, relatively few studies have monitored the fre- by IHC and in situ hybridization, electron microscopy, and
quency of CD4+ and CD8+ T cell subsets into the late stages virus isolation/outgrowth assays. Optimally, biologically rel-
of infection. However, findings from these few studies, evant tissue reservoirs should be identified using a combina-
including experimental and natural infections, show that tion of these techniques.

1989
Eckstrand et al., Journal of General Virology 2017;98:1985–1996

Anatomical reservoirs frequently discussed in the HIV field Multiple reports have shown virus localized to various com-
are the brain, gut-associated lymphoid tissue (GALT), partments of the brain by qPCR or by in situ hybridization
lymph nodes and the reproductive tract [13, 14, 88, 91]. for FIV nucleic acid. However, relatively few in vivo studies
Many of these tissues have been investigated as tissue reser- have tracked specific cell populations within the CNS that
voirs in the FIV-infected cat as well, particularly during the are infected with FIV, with a few reports confirming infec-
acute and early asymptomatic phases of infection [60, 62, tion of resident microglia in experimentally infected cats
79, 92]. A thorough understanding of tissue reservoir distri- [69, 107]. FIV infection of the CNS warrants further scru-
bution and biology requires an understanding of the early tiny to determine the value of this animal model for testing
pathogenesis and dissemination of virus. therapeutic approaches to purge the HIV-1 reservoirs
within the CNS.
FIV inoculation in cats is believed to occur primarily
through blood transmission during fighting (biting), how- There are fewer studies that have examined the long-term
ever many other routes of experimental inoculation have persistence of FIV in tissue sites. However, several studies
proven to be effective for viral transmission, including oral, indicate that tissues targeted by FIV in the acute phase also
rectal/vaginal mucosal transfer, intravenous, intramuscular, harbour replication-competent provirus in the later stages
intraperitoneal, subcutaneous injection and transplacental of infection. Dean et al. demonstrated the presence of repli-
transfer [17, 19, 93–97]. Infection can be achieved by inocu- cation-competent provirus in the peripheral blood and
lation of either cell-associated or cell-free virus, with cell- lymph node out to 70 months (5.8 years) after infection with
free virus being apparently more efficient (at least with FIV [27]. Pistello et al. demonstrated the presence of abun-
FIV-C) [30, 60, 93]. Viral inoculation is followed by robust dant provirus in lymphoid tissues, and lesser amounts in
cell-associated viral replication in the peripheral blood, the brain, lung and kidney 3 years post super-inoculation
resulting a transient plasma viraemia and systemic viral dis- (inoculation followed by second exposure) with FIV-Petal-
semination [60, 81, 98]. Obert and Hoover demonstrated uma [108]. Infection of bone marrow cells in naturally
that within 2 days of oro-nasal or vaginal mucosal exposure, infected asymptomatic cats has also been reported, although
FIV traffics from the mucosa to the regional lymph nodes the identity of infected subsets was not determined [25].
via dendritic cells or CD3+ T cells transfer with subsequent Two recent investigations into tissue reservoirs by our labo-
systemic spread to secondary and tertiary lymphoid organs ratory demonstrated the presence of proviral DNA within
[30]. Other important target tissues in early FIV dissemina- the popliteal lymph node, mesenteric lymph node, spleen
and mucosa of the small intestines during the late asymp-
tion include additional lymphoid organs such as the intesti-
tomatic phase of infection (5–6 years post inoculation), in
nal tract mucosa, thymus, spleen, tonsil and bone marrow
the face of a relatively quiescent or inactive viral harbour
[27, 29, 31, 81, 94, 99–103]. Fig. 1 summarizes important
replication-competent provirus in spite of relatively quies-
tissue and cellular reservoirs known to harbour virus in
cent or inactive viral replication in the peripheral blood [79,
FIV-infected cats.
92, 109]. Interestingly, our studies revealed that, firstly,
Additionally FIV-infected cells can be detected in the CNS, lymph node germinal centres contained an increased fre-
liver, kidney, lung and salivary gland during the acute phase quency of CD3+ T cells compared to uninfected cat tissues.
[74, 81, 94, 97]. Table 1 lists the studies of tissue reservoirs These observations suggested a proliferation of resident of
of FIV. Rogers et al. described a modified IHC technique for Tfh cells or infiltration of CD8+ T cells into the B cell fol-
the detection of FIV protein (undefined FIV antigen) in for- licles, although CD8+ T cells are generally restricted from
malin-fixed, paraffin-embedded tissue by using feline serum entering B cell follicles. Secondly, in our most recent report,
from an experimentally FIV-infected cat [29]. The study a novel in situ hybridization technique (RNAscope) for
demonstrated that, within 3 weeks of inoculation, FIV dis- detection of FIV RNA identified germinal centres in the
seminates to many tissue types including lymph nodes, mesenteric lymph node, spleen and intestinal Peyer’s
spleen, intestinal lamina propria, intestinal Peyer’s patches patches as microanatomical reservoirs of viral transcription
(lymphoid follicles), thymus, bone marrow and resident tis- in the chronically FIV-infected cat [92].
sue histiocytic cells such as Kupffer cells of the liver [29].
Interestingly, brain was not identified as a tissue for FIV FIV LATENCY
infection. However, other reports have demonstrated that Multiple investigative modalities have been used to identify
different FIV strains vary in their tropism for the brain and cellular reservoirs of FIV, including the detection and quan-
for different compartments of the brain [70, 104]. These tification of proviral DNA, viral RNA, viral protein, reverse
findings point to some similarities in viral pathogenesis for transcriptase activity and other techniques described in
the CNS shared by FIV and HIV-1 ([74, 105] – review with detail in a recent review by Ammersbach and Bienzle [110].
references therein). Indeed, based on multiple reports, the While the detection and quantification of these various viral
brain has proven to be an important target during early FIV components can provide supportive evidence of viral repli-
infection and, similar to HIV, has been associated with vari- cation, they do not ultimately prove a cell’s ability to pro-
ous pathologic features such as encephalitis, microgial acti- duce infectious virus. The ‘gold standard assay’ for the
vation and, in some cases, degenerative changes like myelin identification of replication-competent and infectious lenti-
pallor ([74, 105] and references therein; [104, 106]). viral reservoir cells is the ex vivo viral outgrowth assay

1990
Eckstrand et al., Journal of General Virology 2017;98:1985–1996

Cellular reservoirs
Lymphocytes
Tissue reservoirs
CD4+T cells
Lymph nodes CD8+T cells
Bone marrow CD21+B cells
Reproductive tract
Antigen processing/
Gastrointestinal tract presenting cells
Peyer’s patches Monocytes
Spleen Macrophages
Dendritic cells
Thymus
Blood CNS specific
Brain
Microglia
Astrocytes

Fig. 1. Tissue and cellular reservoirs of feline immunodeficiency virus in the infected cat.

(VOA) which has been used extensively for investigation of Lentiviral latency refers to cells with integrated replication-
HIV-1 latency in humans [111] and in our laboratory for competent provirus that are transcriptionally inactive.
detection of cells containing transcriptionally silent infec- Latently infected cells can be identified by detecting proviral
tious provirus in cats chronically infected with FIV [84, 92]. DNA with a concurrent absence of detectable viral RNA
For assessment of virus infection by VOA, an enriched and viral proteins. Lentiviral latency allows the integrated
(purified) cell type such as CD4+ T cells are obtained from and replication-competent provirus to remain essentially
an infected individual and are cultured ex vivo. To reverse invisible to both the host immune system and antiretroviral
potential viral latency, cultured cells are stimulated for vary- therapies and thus allows the virus to persist over time
ing amounts of time with mitogenic compounds such as within the infected cell and host. Latently infected cells are
phytohemagglutinin or concanalin-A and/or allogeneic thus of key importance when considering viral reservoirs,
peripheral blood mononuclear cells (PBMCs) to instigate a particularly within a host on ART. McDonnel et al.
mixed lymphocyte reaction [112]. In the VOA, ex vivo stim- reviewed FIV latency and the strengths of the FIV-infected
ulation is an attempt to ‘reverse latency’ by activating viral cat as a model of the HIV-infected human [61]. Different
transcription, viral protein production, virion assembly and cohorts of naturally and experimentally infected cats have
release from cells containing replication-competent provi- demonstrated either undetectable or consistently detectable
rus. For detection of FIV infection, cell-free (clarified) cul- plasma viraemia during the asymptomatic phase, which
ture supernatant from the activated cells is then passaged may be due to differences in viral strains, inoculating dose,
onto uninfected SPF feline PBMCs or another susceptible age of the animal or other factors [7, 84, 98, 104, 115–118].
cell type. These cells are then cultured, and subsequently The cellular mechanisms involved in FIV latency have been
tested for the presence of proviral DNA by standard or identified in both in vitro and in vivo infection systems [52,
quantitative PCR. Detection of proviral DNA in the second- 84, 119–122]. McDonnel et al. reported that peripheral
ary SPF cell culture confirms that supernatant from the pri- blood-derived CD4+ T cells latently infected with replica-
mary cell culture contained replication-competent and tion-competent, infectious virus were infrequent (estimated
infectious virus. The VOA can also be used to quantify the at approximately 1 cell in 105 peripheral CD4+ T cells). This
size of the reservoir by initially preparing serial dilutions of report also revealed that latent infection was characterized
the reservoir cell of interest [113]. Although this assay is by the association of the FIV 5¢ long terminal repeat
most sensitive for virus detection, major limitations of the (LTR) promoter with host histone proteins that were hyper-
VOA are that ex vivo stimulation may fail to reverse latency methylated and deacetylated, consistent with a condensed
(induce viral transcription) of all the replication-competent chromatin state [123]. Subsequent reports demonstrated
proviruses present within the tested reservoir population, is that latently FIV-infected cells could be reactivated ex vivo
labour intensive and may yield variable results [13]. As a through the administration of histone deacetylase inhibitors
result, apparently ‘non-inducible’ cells harbouring replica- suberoylanilide hydroxamic acid, valproic acid,
tion-competent provirus will fail to be identified [114]. trichostatin A, and sodium butyrate (NaB) and a histone

1991
Eckstrand et al., Journal of General Virology 2017;98:1985–1996

methyltransferase inhibitor 3-deazanoplanocin A [124, inoculating virus concentrations, immune restriction and
125]. Similar findings were reported in two studies by Tada- cell activation, may play a role in FIV latency, factors shared
fumi and Chan who used histone deacytelase NaB and pro- by HIV-1 latency. One report has described FIV proviral
tein kinase-C inhibitors, respectively, to reverse FIV latency DNA integration sites, which were revealed to be similar to
[120, 126]. Lastly, only short, abortive transcripts (R region those for HIV-1 [130]. However, any relationship between
of the LTR) along with an association of RNA polymerase II FIV integration sites and virus latency is currently undeter-
with the LTR by chromosomal immunoprecipitation were mined. Furthermore, in contrast to HIV-1 infection, FIV
detected in cells latently infected with FIV [123]. These latency may be apparent in the absence of ART and a com-
findings provided indirect and direct evidence that RNA mon characteristic in cats naturally infected with FIV, par-
polymerase II activity is paused on the FIV promoter, a ticularly in environments free of stress and exposure to
finding also reported for cells latently infected with HIV other pathogens, such as a single-cat household [7]. How-
[123, 127, 128]. These findings collectively reveal that circu- ever, this dissimilarity suggests that FIV infection may pro-
lating CD4+ T cells serve as a reservoir for latent FIV infec- vide a unique opportunity to examine mechanisms and
tions with latency characterized by at least one similar interventions of lentiviral latency in the absence of ART.
regulatory mechanism, repressive chromatin, that has also
been described for HIV-1 latency in patients on ART [129]. CONCLUSIONS
Assogba et al. suggested that low dose viral inocula (102–103 Since the initial identification of FIV, there has been an
copies), delivered by the mucosal route, and CD8+ T cells extensive history of investigations into cellular and tissue
may also play important roles in inducing latency in CD4+ viral reservoirs of FIV that have greatly contributed to
cells and continued viral suppression [121]. The same study understanding the pathogenesis of disease in the FIV-
described an infection state proposed as latency, in which infected cat. Additionally, there is no shortage of research
proviral DNA was undetectable in blood- and tissue-derived and review papers highlighting the strengths and advantages
cells harvested from experimentally infected cats [121]. of using the FIV-infected cat as an animal model for the
However, activation of cultivated tissue lysates depleted of HIV-infected human. Many tissue reservoirs of FIV are
CD8+ T cells ex vivo resulted in virus production. The iden- very similar to HIV and include (but are not limited to)
tity of cellular subsets serving as reservoirs for latent virus lymphoid tissues, including the gastrointestinal tract, and
within these infected tissues was not determined. A different the CNS. FIV infects similar leukocyte subsets as HIV and
report showed that FIV preferentially replicated in activated most likely utilizes the same cells, such as dendritic cells and
CD4+CD25+ T cells and that CD4+CD25+ T cells served as astrocytes, to facilitate infection of T cell targets. Impor-
a productive reservoir resistant to apoptotic cell death [52]. tantly, FIV infection offers the opportunity to investigate
In contrast, CD4+CD25- cells were shown to be reservoirs the relevance of cellular and tissue reservoirs that may not
for latent FIV with the potential to reactivate. This observa- be readily accessible in HIV-infected people (i.e. CNS). Fur-
tion suggests that less-activated CD4+ T cells, as defined by thermore, examination of the differences between FIV and
the absence of CD25 expression, may preferentially harbour HIV-1 infections, including receptor usage and clinical out-
latent FIV infection – a finding that agrees with the observa- comes, may also provide unique insights to the viral mecha-
nisms of lentiviral pathogenesis. As the lentivirus field
tion of resting CD4+ T cells being a major reservoir for
evolves to investigate new antiretroviral and eradication
latent HIV-1 infection in patients on ART [13]. CD4+ T
strategies such as the gene-editing tool, CRISPR, one should
cells are the most frequently investigated cell type with
consider the advantages of utilizing the affordable, outbred,
regards to FIV latency. However, testing with additional
tractable, naturally occurring FIV cat disease model for test-
markers for feline T cell activation and memory will be nec-
ing the safety and efficacy of such therapies.
essary to fully characterize the CD4+ T cell subsets that are
the predominant reservoirs for latent FIV infection and to
determine similarities between latency FIV and HIV-1 for Funding information
CD4 + T cell infections. Finally, a previous report revealed The authors received no specific grant from any funding agency.
that monocytes isolated 12–16 weeks after experimental FIV
Conflicts of interest
inoculation contained detectable proviral DNA in the The authors declare that there are no conflicts of interest.
absence of detectable viral protein. Moreover, virus produc-
tion from these monocytes could be subsequently induced References
when stimulated ex vivo [58]. Monocytes and macrophages 1. Levy JK, Scott HM, Lachtara JL, Crawford PC. Seroprevalence
of feline leukemia virus and feline immunodeficiency virus
have also been identified as reservoirs for latent HIV-1 infection among cats in North America and risk factors for sero-
infection in patients on ART [13]. Taken together, observa- positivity. J Am Vet Med Assoc 2006;228:371–376.
tions to date suggest that FIV may share similarities in cellu- 2. Nedumpun T, Piamsomboon P, Chanchaithong P,
lar reservoirs and molecular mechanisms for latency with Taweethavonsawat P, Chungpivat S et al. Prevalence and distribu-
tions of feline immunodeficiency virus and feline leukemia virus
HIV-1, although FIV latency remains less well characterized infections in Bangkok and its vicinity, Thailand during 2013–2014.
compared to HIV-1 latency, particularly regarding mecha- THAI J Vet Med 2015;45:449–453.
nisms of latency. These few reports described for FIV sug- 3. Oguzoglu T, Muz D, Timurkan M, Maral N, Gurcan I. Prevalences
gest that epigenetic factors, early virus infection events such of feline coronavirus (FCoV), feline leukaemia virus (FeLV), feline

1992
Eckstrand et al., Journal of General Virology 2017;98:1985–1996

immunodeficiency virus (FIV) and feline parvovirus (FPV) among 22. Naif HM. Pathogenesis of HIV infection. Infect Dis Rep 2013;5:
domestic cats in Ankara, Turkey. Rev Med Vet 2013;164:511– 26–30.
516. 23. Ishida T, Tomoda I. Clinical staging of feline immunodeficiency
4. Bande F, Arshad SS, Hassan L, Zakaria Z, Sapian NA et al. virus infection. Japanese J Vet Sci 1990;52:645–648.
Prevalence and risk factors of feline leukaemia virus and feline 24. Hartmann K. Clinical aspects of feline retroviruses: a review.
immunodeficiency virus in peninsular Malaysia. BMC Vet Res Viruses 2012;4:2684–2710.
2012;8:33.
25. Fujino Y, Horiuchi H, Mizukoshi F, Baba K, Goto-Koshino Y et al.
5. Nakamura Y, Nakamura Y, Ura A, Hirata M, Sakuma M et al. An
Prevalence of hematological abnormalities and detection of
updated nation-wide epidemiological survey of feline immuno-
infected bone marrow cells in asymptomatic cats with feline
deficiency virus (FIV) infection in Japan. J Vet Med Sci 2010;72:
immunodeficiency virus infection. Vet Microbiol 2009;136:217–
1051–1056.
225.
6. Hartmann K. Clinical aspects of feline immunodeficiency and
26. English RV, Johnson CM, Gebhard DH, Tompkins MB. In vivo
feline leukemia virus infection. Vet Immunol Immunopathol 2011;
143:190–201. lymphocyte tropism of feline immunodeficiency virus. J Virol
1993;67:5175–5186.
7. Bęczkowski PM, Litster A, Lin TL, Mellor DJ, Willett BJ et al.
Contrasting clinical outcomes in two cohorts of cats naturally 27. Dean GA, Reubel GH, Moore PF, Pedersen NC. Proviral burden
infected with feline immunodeficiency virus (FIV). Vet Microbiol and infection kinetics of feline immunodeficiency virus in lym-
2015;176:50–60. phocyte subsets of blood and lymph node. J Virol 1996;70:5165–
5169.
8. Pedersen NC, Torten M, Rideout B, Sparger E, Tonachini T et al.
Feline leukemia virus infection as a potentiating cofactor for the 28. Dean GA, Himathongkham S, Sparger EE. Differential cell tro-
primary and secondary stages of experimentally induced feline pism of feline immunodeficiency virus molecular clones in vivo.
immunodeficiency virus infection. J Virol 1990;64:598–606. J Virol 1999;73:2596-603.
9. Kohmoto M, Uetsuka K, Ikeda Y, Inoshima Y, Shimojima M et al. 29. Rogers AB, Mathiason CK, Hoover EA. Immunohistochemical
Eight-year observation and comparative study of specific patho- localization of feline immunodeficiency virus using native spe-
gen-free cats experimentally infected with feline immunodefi- cies antibodies. Am J Pathol 2002;161:1143–1151.
ciency virus (FIV) subtypes A and B: terminal acquired 30. Obert LA, Hoover EA. Early pathogenesis of transmucosal feline
immunodeficiency syndrome in a cat infected with FIV petaluma immunodeficiency virus infection. J Virol 2002;76:6311–6322.
strain. J Vet Med Sci 1998;60:315–321.
31. Beebe AM, Dua N, Faith TG, Moore PF, Pedersen NC et al. Pri-
10. Kann RK, Seddon JM, Kyaw-Tanner MT, Henning J, Meers J. mary stage of feline immunodeficiency virus infection: viral dis-
Association between feline immunodeficiency virus (FIV) plasma semination and cellular targets. J Virol 1994;68:3080–3091.
viral RNA load, concentration of acute phase proteins and dis-
32. Willett BJ, Picard L, Hosie MJ, Turner JD, Adema K et al.
ease severity. Vet J 2014;201:181–183.
Shared usage of the chemokine receptor CXCR4 by the feline
11. Yoshikawa R, Izumi T, Yamada E, Nakano Y, Misawa N et al. A and human immunodeficiency viruses. J Virol 1997;71:6407–
naturally occurring domestic cat APOBEC3 variant confers 6415.
resistance to feline immunodeficiency virus infection. J Virol
2015;90:474–485. 33. de Parseval A, Chatterji U, Sun P, Elder JH. Feline immunodefi-
ciency virus targets activated CD4+ T cells by using CD134 as a
12. Kolenda-Roberts HM, Kuhnt LA, Jennings RN, Mergia A, binding receptor. Proc Natl Acad Sci USA 2004;101:13044–
Gengozian N et al. Immunopathogenesis of feline immunodefi- 13049.
ciency virus infection in the fetal and neonatal cat. Front Biosci
2007;12:3668–3682. 34. Shimojima M, Miyazawa T, Ikeda Y, Mcmonagle EL, Haining H
et al. Use of CD134 as a primary receptor by the feline immuno-
13. Barton K, Winckelmann A, Palmer S. HIV-1 reservoirs during
deficiency virus. Science 2004;303:1192–1195.
suppressive therapy. Trends Microbiol 2016;24:345–355.
35. de Parseval A, Chatterji U, Morris G, Sun P, Olson AJ et al.
14. Churchill MJ, Deeks SG, Margolis DM, Siliciano RF, Swanstrom
Structural mapping of CD134 residues critical for interaction
R. HIV reservoirs: what, where and how to target them. Nat Rev
with feline immunodeficiency virus. Nat Struct Mol Biol 2005;12:
Microbiol 2016;14:55–60.
60–66.
15. Chavez L, Calvanese V, Verdin E. HIV latency is established
36. Hu QY, Fink E, Hong Y, Wang C, Grant CK et al. Fine definition of
directly and early in both resting and activated primary CD4 T
cells. PLoS Pathog 2015;11:e1004955. the CXCR4-binding region on the V3 loop of feline immunodefi-
ciency virus surface glycoprotein. PLoS One 2010;5:e10689.
16. Morrison JH, Guevara RB, Marcano AC, Saenz DT, Fadel HJ
et al. Feline immunodeficiency virus envelope glycoproteins 37. Aiamkitsumrit B, Dampier W, Antell G, Rivera N, Martin-Garcia
antagonize tetherin through a distinctive mechanism that J et al. Bioinformatic analysis of HIV-1 entry and pathogenesis.
requires virion incorporation. J Virol 2014;88:3255–3272. Curr HIV Res 2014;12:132–161.
17. Pedersen NC, Ho EW, Brown ML, Yamamoto JK. Isolation of a 38. Clapham PR, Mcknight A. Cell surface receptors, virus entry
T-lymphotropic virus from domestic cats with an immunodefi- and tropism of primate lentiviruses. J Gen Virol 2002;83:1809–
ciency-like syndrome. Science 1987;235:790–793. 1829.
18. Hopper CD, Sparkes AH, Gruffydd-Jones TJ, Crispin SM, Muir P 39. Croft M. Control of immunity by the TNFR-related molecule
et al. Clinical and laboratory findings in cats infected with feline OX40 (CD134). Annu Rev Immunol 2010;28:57–78.
immunodeficiency virus. Vet Rec 1989;125:341–346. 40. Aspeslagh S, Postel-Vinay S, Rusakiewicz S, Soria JC, Zitvogel
19. Torten M, Franchini M, Barlough JE, George JW, Mozes E et al. L et al. Rationale for anti-OX40 cancer immunotherapy. Eur J
Progressive immune dysfunction in cats experimentally infected Cancer 2016;52:50–66.
with feline immunodeficiency virus. J Virol 1991;65:2225–2230. 41. Tahiliani V, Hutchinson TE, Abboud G, Croft M, Salek-Ardakani
20. English RV, Nelson P, Johnson CM, Nasisse M, Tompkins WA S. OX40 cooperates with ICOS to amplify follicular th cell devel-
et al. Development of clinical disease in cats experimentally opment and germinal Center reactions during infection.
infected with feline immunodeficiency virus. J Infect Dis 1994; J Immunol 2017;198:218–228.
170:543–552. 42. Willett BJ, Mcmonagle EL, Logan N, Spiller OB, Schneider P
21. Elder JH, Lin YC, Fink E, Grant CK. Feline immunodeficiency et al. Probing the interaction between feline immunodeficiency
virus (FIV) as a model for study of lentivirus infections: parallels virus and CD134 by using the novel monoclonal antibody 7D6
with HIV. Curr HIV Res 2010;8:73–80. and the CD134 (Ox40) ligand. J Virol 2007;81:9665–9679.

1993
Eckstrand et al., Journal of General Virology 2017;98:1985–1996

43. Reggeti F, Ackerley C, Bienzle D. CD134 and CXCR4 expression 63. Hood S, Thompson E, Akaronu N, Miller M, Virology FJ. Mono-
corresponds to feline immunodeficiency virus infection of lym- cyte-derived dendritic cells from feline immunodeficiency virus
phocytes, macrophages and dendritic cells. J Gen Virol 2008;89: positive cats are productively infected and maintain CD8+ T cell
277–287. stimulatory capacity. Int J Virol AIDS 2015;2:1.
44. Willett BJ, Mcmonagle EL, Ridha S, Hosie MJ. Differential utili- 64. Sprague WS, Robbiani M, Avery PR, O’Halloran KP, Hoover EA.
zation of CD134 as a functional receptor by diverse strains of Feline immunodeficiency virus dendritic cell infection and trans-
feline immunodeficiency virus. J Virol 2006;80:3386–3394. fer. J Gen Virol 2008;89:709–715.
45. Willett BJ, Mcmonagle EL, Bonci F, Pistello M, Hosie MJ. Map- 65. Nasr N, Harman A, Turville S, Cunningham AL. HIV infection of
ping the domains of CD134 as a functional receptor for feline dendritic cells. Methods Mol Biol 2014;1087:221–232.
immunodeficiency virus. J Virol 2006;80:7744–7747. 66. Miles B, Connick E. TFH in HIV latency and as sources of repli-
46. Grant CK, Fink EA, Sundstrom M, Torbett BE, Elder JH. cation-competent virus. Trends Microbiol 2016;24:338–344.
Improved health and survival of FIV-infected cats is associated 67. Dow SW, Dreitz MJ, Hoover EA. Feline immunodeficiency virus
with the presence of autoantibodies to the primary receptor, neurotropism: evidence that astrocytes and microglia are the
CD134. Proc Natl Acad Sci USA 2009;106:19980–19985. primary target cells. Vet Immunol Immunopathol 1992;35:23–35.
47. Bęczkowski PM, Techakriengkrai N, Logan N, Mcmonagle E, 68. Dow SW, Poss ML, Hoover EA. Feline immunodeficiency virus: a
Litster A et al. Emergence of CD134 cysteine-rich domain 2 neurotropic lentivirus. J Acquir Immune Defic Syndr 1990;3:658–
(CRD2)-independent strains of feline immunodeficiency virus 668.
(FIV) is associated with disease progression in naturally infected
cats. Retrovirology 2014;11:95. 69. Hein A, Martin JP, Koehren F, Bingen A, Dörries R. In vivo infec-
tion of ramified microglia from adult cat central nervous system
48. Bęczkowski PM, Hughes J, Biek R, Litster A, Willett BJ et al. by feline immunodeficiency virus. Virology 2000;268:420–429.
Feline immunodeficiency virus (FIV) env recombinants are com-
mon in natural infections. Retrovirology 2014;11:80. 70. Billaud JN, Selway D, Yu N, Phillips TR. Replication rate of feline
immunodeficiency virus in astrocytes is envelope dependent:
49. Willett BJ, Hosie MJ. The virus-receptor interaction in the repli- implications for glutamate uptake. Virology 2000;266:180–188.
cation of feline immunodeficiency virus (FIV). Curr Opin Virol
2013;3:670–675. 71. Yu N, Billaud JN, Phillips TR. Effects of feline immunodeficiency
virus on astrocyte glutamate uptake: implications for lentivirus-
50. Willett BJ, Kraase M, Logan N, Mcmonagle E, Varela M et al.
induced central nervous system diseases. Proc Natl Acad Sci
Selective expansion of viral variants following experimental
USA 1998;95:2624–2629.
transmission of a reconstituted feline immunodeficiency virus
quasispecies. PLoS One 2013;8:e54871. 72. Ryan G, Grimes T, Brankin B, Mabruk MJ, Hosie MJ et al. Neu-
ropathology associated with feline immunodeficiency virus
51. Willett BJ, Cannon CA, Hosie MJ. Expression of CXCR4 on feline
infection highlights prominent lymphocyte trafficking through
peripheral blood mononuclear cells: effect of feline immunodefi-
both the blood-brain and blood-choroid plexus barriers.
ciency virus infection. J Virol 2003;77:709–712.
J Neurovirol 2005;11:337–345.
52. Joshi A, Vahlenkamp TW, Garg H, Tompkins WA, Tompkins MB.
73. Meeker RB, Bragg DC, Poulton W, Hudson L. Transmigration of
Preferential replication of FIV in activated CD4+ CD25+ T cells
macrophages across the choroid plexus epithelium in response
independent of cellular proliferation. Virology 2004;321:307–
to the feline immunodeficiency virus. Cell Tissue Res 2012;347:
322.
443–455.
53. Hu QY, Fink E, Elder JH. Mapping of receptor binding interac-
74. Fletcher NF, Meeker RB, Hudson LC, Callanan JJ. The neuropa-
tions with the FIV surface glycoprotein (SU); Implications
thogenesis of feline immunodeficiency virus infection: barriers
regarding immune surveillance and cellular targets of infection.
to overcome. Vet J 2011;188:260–269.
Retrovirology 2012;2012:1–11.
75. Gorry PR, Ong C, Thorpe J, Bannwarth S, Thompson KA et al.
54. Troth SP, Dean AD, Hoover EA. In vivo CXCR4 expression, lym-
Astrocyte infection by HIV-1: mechanisms of restricted virus
phoid cell phenotype, and feline immunodeficiency virus infec-
replication, and role in the pathogenesis of HIV-1-associated
tion. Vet Immunol Immunopathol 2008;123:97–105.
dementia. Curr HIV Res 2003;1:463–473.
55. Bobardt MD, Saphire AC, Hung HC, Yu X, van der Schueren B
76. Liu Y, Liu H, Kim BO, Gattone VH, Li J et al. CD4-independent
et al. Syndecan captures, protects, and transmits HIV to T lym-
infection of astrocytes by human immunodeficiency virus type
phocytes. Immunity 2003;18:27–39.
1: requirement for the human mannose receptor. J Virol 2004;
56. Geijtenbeek TB, Kwon DS, Torensma R, van Vliet SJ, van 78:4120–4133.
Duijnhoven GC et al. DC-SIGN, a dendritic cell-specific HIV-1-
77. Gray LR, Turville SG, Hitchen TL, Cheng WJ, Ellett AM et al.
binding protein that enhances trans-infection of T cells. Cell
HIV-1 entry and trans-infection of astrocytes involves CD81
2000;100:587–597.
vesicles. PLoS One 2014;9:e90620.
57. Woo JC, Dean GA, Pedersen NC, Moore PF. Immunopathologic
78. Russell RA, Chojnacki J, Jones DM, Johnson E, Do T et al.
changes in the thymus during the acute stage of experimentally
Astrocytes resist HIV-1 fusion but Engulf infected macrophage
induced feline immunodeficiency virus infection in juvenile cats.
material. Cell Rep 2017;18:1473–1483.
J Virol 1997;71:8632–8641.
79. Eckstrand CD, Hillman C, Smith AL, Sparger EE, Murphy BG.
58. Dow SW, Mathiason CK, Hoover EA, Edward A. In vivo monocyte
Viral reservoirs in lymph nodes of FIV-infected progressor and
tropism of pathogenic feline immunodeficiency viruses. J Virol
long-term non-progressor cats during the asymptomatic phase.
1999;73:6852–6861.
PLoS One 2016;11:e0146285.
59. Brown WC, Bissey L, Logan KS, Pedersen NC, Elder JH et al.
80. Willett BJ, Hosie MJ, Callanan JJ, Neil JC, Jarrett O. Infection
Feline immunodeficiency virus infects both CD4+ and CD8+ T
lymphocytes. J Virol 1991;65:3359–3364. with feline immunodeficiency virus is followed by the rapid
expansion of a CD8+ lymphocyte subset. Immunology 1993;78:
60. Burkhard MJ, Dean GA. Transmission and immunopathogenesis 1–6.
of FIV in cats as a model for HIV. Curr HIV Res 2003;1:15–29.
81. Dua N, Reubel G, Moore PF, Higgins J, Pedersen NC. An experi-
61. Mcdonnel SJ, Sparger EE, Murphy BG. Feline immunodeficiency mental study of primary feline immunodeficiency virus infection
virus latency. Retrovirology 2013;10:69. in cats and a historical comparison to acute simian and human
62. Bienzle D. FIV in cats–a useful model of HIV in people? Vet immunodeficiency virus diseases. Vet Immunol Immunopathol
Immunol Immunopathol 2014;159:171–179. 1994;43:337–355.

1994
Eckstrand et al., Journal of General Virology 2017;98:1985–1996

82. Sprague WS, Terwee JA, Vandewoude S. Temporal association immunodeficiency virus infected cats. Arch Virol 2002;147:
of large granular lymphocytosis, neutropenia, proviral load, and 1053–1059.
FasL mRNA in cats with acute feline immunodeficiency virus 101. Thompson J, Macmillan M, Boegler K, Wood C, Elder JH et al.
infection. Vet Immunol Immunopathol 2010;134:115–121. Pathogenicity and rapid growth kinetics of feline immunodefi-
83. Murphy B, Hillman C, Mcdonnel S. Peripheral immunophenotype ciency virus are linked to 3’ elements. PLoS One 2011;6:e24020.
and viral promoter variants during the asymptomatic phase of 102. Tanabe T, Yamamoto JK. Phenotypic and functional characteris-
feline immunodeficiency virus infection. Virus Res 2014;179:34– tics of FIV infection in the bone marrow stroma. Virology 2001;
43. 282:113–122.
84. Murphy B, Vapniarsky N, Hillman C, Castillo D, Mcdonnel S 103. Beebe AM, Gluckstern TG, George J, Pedersen NC, Dandekar S.
et al. FIV establishes a latent infection in feline peripheral blood Detection of feline immunodeficiency virus infection in bone
CD4+ T lymphocytes in vivo during the asymptomatic phase of marrow of cats. Vet Immunol Immunopathol 1992;35:37–49.
infection. Retrovirology 2012;9:12–17.
104. Miller C, Bielefeldt-Ohmann H, Macmillan M, Huitron-Resendiz
85. Bingen A, Nonnenmacher H, Bastien-Valle M, Martin JP. Tis-
S, Henriksen S et al. Strain-specific viral distribution and neuro-
sues rich in macrophagic cells are the major sites of feline
pathology of feline immunodeficiency virus. Vet Immunol
immunodeficiency virus uptake after intravenous inoculation
Immunopathol 2011;143:282–291.
into cats. Microbes Infect 2002;4:795–803.
105. Calcagno A, Di Perri G, Bonora S. Treating HIV infection in the
86. Queen SE, Mears BM, Kelly KM, Dorsey JL, Liao Z et al. Replica-
central nervous system. Drugs 2017;77:145–157.
tion-competent simian immunodeficiency virus (SIV) Gag escape
mutations archived in latent reservoirs during antiretroviral 106. Rao VR, Ruiz AP, Prasad VR. Viral and cellular factors underly-
treatment of SIV-infected macaques. J Virol 2011;85:9167–9175. ing neuropathogenesis in HIV associated neurocognitive disor-
ders (HAND). AIDS Res Ther 2014;11:13.
87. North TW, Higgins J, Deere JD, Hayes TL, Villalobos A et al.
Viral sanctuaries during highly active antiretroviral therapy in a 107. Hein A, Martin JP, Dörries R. Early pathological changes in the
nonhuman primate model for AIDS. J Virol 2010;84:2913–2922. central nervous system of acutely feline-immunodeficiency-
virus-infected cats. Virology 2005;343:162–170.
88. Eisele E, Siliciano RF. Redefining the viral reservoirs that pre-
vent HIV-1 eradication. Immunity 2012;37:377–388. 108. Pistello M, Matteucci D, Cammarota G, Mazzetti P, Giannecchini
89. Letendre S, Marquie-Beck J, Capparelli E, Best B, Clifford D S et al. Kinetics of replication of a partially attenuated virus and
et al. Validation of the CNS penetration-effectiveness rank for of the challenge virus during a three-year intersubtype feline
quantifying antiretroviral penetration into the central nervous immunodeficiency virus superinfection experiment in cats.
system. Arch Neurol 2008;65:65–70. J Virol 1999;73:1518–1527.
90. Fletcher CV, Staskus K, Wietgrefe SW, Rothenberger M, Reilly 109. Eckstrand C, Hillman C, Murphy B. Sequence instability in the
C et al. Persistent HIV-1 replication is associated with lower proviral long terminal repeat and gag regions from peripheral
antiretroviral drug concentrations in lymphatic tissues. Proc blood and tissue-derived leukocytes of FIV-infected cats during
Natl Acad Sci USA 2014;111:2307–2312. the late asymptomatic phase. Vet Sci 2016;3:10.
91. Svicher V, Ceccherini-Silberstein F, Antinori A, Aquaro S, Perno 110. Ammersbach M, Bienzle D. Methods for assessing feline immu-
CF et al. Understanding HIV compartments and reservoirs. Curr nodeficiency virus infection, infectivity and purification. Vet
HIV/AIDS Rep 2014;11:186–194. Immunol Immunopathol 2011;143:202–214.
92. Eckstrand CD, Sparger EE, Pitt KA, Murphy BG. Peripheral and 111. Bruner KM, Hosmane NN, Siliciano RF. Towards an HIV-1 cure:
central immune cell reservoirs in tissues from asymptomatic measuring the latent reservoir. Trends Microbiol 2015;23:192–
cats chronically infected with feline immunodeficiency virus. 203.
PLoS One 2017;12:e0175327. 112. Siliciano JD, Siliciano RF. Enhanced Culture Assay for Detection
93. Obert LA, Hoover EA. Feline immunodeficiency virus clade C and Quantitation of Latently Infected, Resting Virus in HIV-1-
mucosal transmission and disease courses. AIDS Res Hum Infected Individuals. In: Zhu T (editor). Methods in Molecular
Retroviruses 2000;16:677–688. Biology. Totowa, NJ: Humana Press; 2005. pp. 3–15.
94. Rogers AB, Hoover EA. Maternal-fetal feline immunodeficiency 113. Laird GM, Eisele EE, Rabi SA, Lai J, Chioma S et al. Rapid quan-
virus transmission: timing and tissue tropisms. J Infect Dis tification of the latent reservoir for HIV-1 using a viral outgrowth
1998;178:960–967. assay. PLoS Pathog 2013;9:e1003398.
95. Burkhard MJ, Obert LA, O’Neil LL, Diehl LJ, Hoover EA. Mucosal 114. Ho YC, Shan L, Hosmane NN, Wang J, Laskey SB et al. Replica-
transmission of cell-associated and cell-free feline immunodefi- tion-competent noninduced proviruses in the latent reservoir
ciency virus. AIDS Res Hum Retroviruses 1997;13:347–355. increase barrier to HIV-1 cure. Cell 2013;155:540–551.
96. Murphy B, Hillman C, Mok M, Vapniarsky N. Lentiviral latency in 115. Miller MM, Fogle JE. Administration of Fozivudine tidoxil as a
peripheral CD4+ T cells isolated from feline immunodeficiency single-agent therapeutic during acute feline immunodeficiency
virus-infected cats during the asymptomatic phase is not asso- virus infection does not alter chronic infection. Viruses 2012;4:
ciated with hypermethylation of the proviral promoter. Virus Res 954–962.
2012;169:117–126. 116. Freer G, Matteucci D, Mazzetti P, Tarabella F, Ricci E et al.
97. Park HS, Kyaw-Tanner M, Thomas J, Robinson WF. Feline Immunotherapy with internally inactivated virus loaded dendritic
immunodeficiency virus replicates in salivary gland ductular cells boosts cellular immunity but does not affect feline immu-
epithelium during the initial phase of infection. Vet Microbiol nodeficiency virus infection course. Retrovirology 2008;5:33.
1995;46:257–267. 117. Kraase M, Sloan R, Klein D, Logan N, Mcmonagle L et al. Feline
98. Diehl LJ, Mathiason-Dubard CK, O’Neil LL, Hoover EA. Longitu- immunodeficiency virus env gene evolution in experimentally
dinal assessment of feline immunodeficiency virus kinetics in infected cats. Vet Immunol Immunopathol 2010;134:96–106.
plasma by use of a quantitative competitive reverse transcrip- 118. Pecon-Slattery J, Troyer JL, Johnson WE, O’Brien SJ. Evolution
tase PCR. J Virol 1995;69:2328–2332. of feline immunodeficiency virus in Felidae: implications for
99. Toyosaki T, Miyazawa T, Furuya T, Tomonaga K, Shin YS et al. human health and wildlife ecology. Vet Immunol Immunopathol
Localization of the viral antigen of feline immunodeficiency virus 2008;123:32–44.
in the lymph nodes of cats at the early stage of infection. Arch 119. Ikeda Y, Tomonaga K, Kawaguchi Y, Kohmoto M, Inoshima Y
Virol 1993;131:335–347. et al. Feline immunodeficiency virus can infect a human cell line
100. Sandy JR, Robinson WF, Bredhauer B, Kyaw-Tanner M, Howlett (MOLT-4) but establishes a state of latency in the cells. J Gen
CR. Productive infection of the bone marrow cells in feline Virol 1996;77:1623–1630.

1995
Eckstrand et al., Journal of General Virology 2017;98:1985–1996

120. Tochikura TS, Naito Y, Kozutsumi Y, Hohdatsu T. Induction 125. Mcdonnel SJ, Sparger EE, Luciw PA, Murphy BG. Pharmaco-
of feline immunodeficiency virus from a chronically infected logic reactivation of latent feline immunodeficiency virus ex vivo
feline T-lymphocyte cell line. Res Vet Sci 2012;92:327–332. in peripheral CD4+ T-lymphocytes. Virus Res 2012;170:174–179.
121. Assogba BD, Leavell S, Porter K, Burkhard MJ. Mucosal 126. Chan CN, Mcmonagle EL, Hosie MJ, Willett BJ. Prostratin exhib-
administration of low-dose cell-associated feline immunodefi- its both replication enhancing and inhibiting effects on FIV infec-
ciency virus promotes viral latency. J Infect Dis 2007;195: tion of feline CD4+ T-cells. Virus Res 2013;171:121–128.
1184–1188. 127. Lenasi T, Contreras X, Peterlin BM. Transcriptional interference
antagonizes proviral gene expression to promote HIV latency.
122. Tomonaga K, Inoshima Y, Ikeda Y, Mikami T. Temporal patterns
Cell Host Microbe 2008;4:123–133.
of feline immunodeficiency virus transcripts in peripheral blood
cells during the latent stage of infection. J Gen Virol 1995;76: 128. Zhang Z, Klatt A, Gilmour DS, Henderson AJ. Negative elonga-
2193–2204. tion factor NELF represses human immunodeficiency virus
transcription by pausing the RNA polymerase II complex. J Biol
123. Mcdonnel SJ, Sparger EE, Luciw PA, Murphy BG. Transcrip- Chem 2007;282:16981–16988.
tional regulation of latent feline immunodeficiency virus in
peripheral CD4+ T-lymphocytes. Viruses 2012;4:878–888. 129. Hakre S, Chavez L, Shirakawa K, Verdin E. Epigenetic regula-
tion of HIV latency. Curr Opin HIV AIDS 2011;6:19–24.
124. Mcdonnel SJ, Liepnieks ML, Murphy BG. Treatment of chroni-
130. Kang Y, Moressi CJ, Scheetz TE, Xie L, Tran DT et al. Integration
cally FIV-infected cats with suberoylanilide hydroxamic acid. site choice of a feline immunodeficiency virus vector. J Virol
Antiviral Res 2014;108:74–78. 2006;80:8820–8823.

Five reasons to publish your next article with a Microbiology Society journal
1. The Microbiology Society is a not-for-profit organization.
2. We offer fast and rigorous peer review – average time to first decision is 4–6 weeks.
3. Our journals have a global readership with subscriptions held in research institutions around
the world.
4. 80% of our authors rate our submission process as ‘excellent’ or ‘very good’.
5. Your article will be published on an interactive journal platform with advanced metrics.

Find out more and submit your article at microbiologyresearch.org.

1996

You might also like