You are on page 1of 7

Available online at www.sciencedirect.

com

ScienceDirect

Measles pathogenesis, immune suppression and animal


models
Brigitta M Laksono1, Rory D de Vries1, W Paul Duprex2,3 and
Rik L de Swart1

Measles virus causes a disease with seemingly innocent appear on the buccal mucosa. The symptomatic phase
symptoms, such as fever and rash. However, measles immune follows a few days later with the appearance of an ery-
suppression causes increased susceptibility to opportunistic thematous, morbilliform skin rash. A robust, virus-specific
infections that are responsible for the majority of over immune response is induced during this phase, which
100 000 yearly fatalities. The pathogenesis of measles is leads to viral clearance, recovery from symptoms and
complex, because measles virus uses multiple receptors to lifelong immunity to measles. Measles also paradoxically
infect different cell types in different phases of the disease. induces a transient immune suppression, which leads to
Experimental morbillivirus infections with wild-type viruses in increased levels of morbidity and mortality up to several
natural host species have demonstrated that direct infection years after recovery [2,3]. The course of measles is
and depletion of memory immune cells causes immune illustrated in Figure 1. MV remains a major global health
amnesia. This was confirmed in studies of a measles outbreak threats to humans, causing more than 140 000 deaths in
in unvaccinated children and provides an explanation for 2018 [4].
epidemiological observations of long-term increases in
morbidity and mortality after measles. To study the pathogenesis of measles, it is crucial to
understand the interaction between MV and its target
Addresses
1
cells. MV infects cells by releasing its encapsidated
Department of Viroscience, Erasmus MC, Dr. Molewaterplein 40, negative-sensed genome into the host cell after recep-
3015 GD Rotterdam, The Netherlands
2
Centre for Vaccine Research, University of Pittsburgh School of tor-dependent virus-to-cell membrane fusion [5]. CD150
Medicine, 3501 Fifth Avenue, Pittsburgh, PA15261, United States (also known as signalling lymphocyte activation mole-
3
Department of Microbiology and Molecular Genetics, University of cule family member 1 or SLAMF1) and nectin-4 (also
Pittsburgh School of Medicine, 3501 Fifth Avenue, Pittsburgh, PA15261, known as poliovirus receptor related-4 or PVRL-4) are
United States
the cellular receptors of wild-type MV, which explain
Corresponding author: de Swart, Rik L (r.deswart@erasmusmc.nl) virus lymphotropism and epitheliotropism, respectively
[6,7,8,9]. CD150 is expressed by several subsets of
immune cells, including macrophages, dendritic cells
Current Opinion in Virology 2020, 41:31–37
(DCs) and lymphocytes. Nectin-4 is a part of the adhe-
This review comes from a themed issue on Special section: measles rens junction complex and expressed on the basolateral
Edited by Rik L de Swart and Makoto Takeda side of polarised epithelial cells. Nectin-4 is also
For a complete overview see the Issue and the Editorial expressed by endothelial cells and keratinocytes
[10,11]. Although the first MV cellular receptor discov-
Available online 24th April 2020
ered was CD46, a cell surface molecule ubiquitously
https://doi.org/10.1016/j.coviro.2020.03.002
expressed by most nucleated cells, subsequent studies
1879-6257/ã 2020 Elsevier B.V. All rights reserved. showed that the CD46-mediated entry is limited to
vaccine and laboratory-adapted MV strains [12,13]. In
addition to these formal entry receptors, MV can also
attach to binding receptors, such as the C-type lectins
DC-specific intercellular adhesion molecule-3-grabbing
non-integrin (DC-SIGN) and Langerin, which are
Introduction expressed on DCs and Langerhans cells, respectively.
Measles virus (MV) is the prototypic member of the Capture of MV particles by these molecules leads to
genus Morbillivirus, family Paramyxoviridae [1], and CD150-mediated infection of DCs (in cis) or lympho-
causes measles in humans and non-human primates cytes (in trans) [14,15]. The ability of MV to infect
(NHPs). In humans, the disease generally occurs during different, highly motile cells in various anatomical com-
childhood and presents after a relatively long incubation partments, such as the respiratory tract, lymphoid tissues,
period. The first signs appear around ten days after gastrointestinal tract and skin, complicates the study of
infection, as fever and cough develop and the patient measles pathogenesis. This review features an integrated
becomes highly contagious. At this point, ulcerated white perspective on the current studies of pathogenesis of
lesions, known as the pathognomonic Koplik’s spots, measles and measles-associated immune suppression.

www.sciencedirect.com Current Opinion in Virology 2020, 41:31–37


32 Measles

Figure 1

Pathogenesis of measles
Infection of local Systemic Immune
Entry Prodromal Symptomatic Recovery
lymphnodes dissemination suppression

O. Composition of lymphocyte population before and after measles


Newly expanded lympocytes

Pre-existing CD150+
MV-specific lymphocytes
lymphocytes

Pre-existing CD150-
lymphocytes

P.Type of cells infected by MV over time

Non-ciliated Ciliated
DC Macrophage Lymphocyte Keratinocyte MV RNP
epithelial cell epithelial cell

Current Opinion in Virology

The pathogenesis of measles. Schematic representation of the course of MV infection. The green bell-shaped curve in the background of the top
panel represents the viral load over time. MV enters the upper (a) or lower respiratory tract (b) by infecting CD150+ or binding to DC-SIGN+
dendritic cells (DCs) or alveolar macrophages. These infected cells migrate to bronchus-associated lymphoid tissues (c) or draining lymph nodes
(d) to spread the infection further to susceptible lymphocytes. MV-infected CD150+ lymphocytes, mostly CD4+ memory T cells, disseminate the

Current Opinion in Virology 2020, 41:31–37 www.sciencedirect.com


Measles pathogenesis, immune suppression and animal models Laksono et al. 33

Animal models andpathologicaloutcomestothoseinhumansandthus have


Mice are not naturally susceptible to MV infection, been commonly used as an animal model for measles [22].
necessitating the development of transgenic mouse mod- Despite the similarities, the two macaque species still differ
els to study measles pathogenesis. However, most of in their clinical presentation. Rhesus macaques have been
these mice are receptor knock-in animals deficient in reported to develop more distinct skin rash and conjuncti-
type I interferon signalling to facilitate wild-type MV vitis than cynomolgus macaques [22–24]. The use of NHPs
infection and replication. Thus, data obtained from these as laboratory animals also raises ethical concerns and is
models should be interpreted cautiously. Infections with progressively more strictly regulated. Nonetheless, the
closely related animal morbilliviruses can be studied in availability of cross-reactive reagents makes macaques
their natural hosts as an alternative, for example canine the ideal animal for in vivo measles studies. Furthermore,
distemper virus (CDV) is a useful surrogate to study the the use of recombinant MVs (rMVs) based on wild-type
pathogenesis of MV infections. CDV infects a broad strains expressing a fluorescent reporter protein allows the
range of hosts, such as dogs, raccoons and ferrets thorough observation of viral tropism, disease kinetics and
[16,17]. Among these hosts, ferrets (Mustela putorius furo) pathogenesis. Altogether, a good animal model for measles
can be handled in a laboratory environment and have a requires the right selection of virus, the susceptibility of the
respiratory tract similar to humans. Recombinant CDV host and the availability of tools and methods to follow the
(rCDV) strains expressing a fluorescent reporter protein course of the disease.
have been used in ferret models, allowing sensitive
detection of infected cells and tissues and determination
of morbilliviral tropism in its natural host [16]. Lastly, the MV entry
use of ferrets as an animal model for other viral diseases The highly infectious MV is transmitted by aerosols and
has also been established and, although limited, reagents small droplets. Respiratory epithelial cells are not the
and tools are available [18]. Despite these advantages, initial targets of the virus, because nectin-4 is only
wild-type CDV in ferrets is not an ideal mimic of MV expressed at the basolateral side [25]. Inoculation of
infection in primates, since CDV infection in ferrets NHPs with wild-type rMV expressing enhanced green
typically results in faster disease progression, neurologi- fluorescent protein showed that myeloid cells in the
cal complications and high mortality rates, while MV respiratory submucosa and the lungs are the initial
infection in primates rarely results in neurological com- target cells in an MV infection, suggesting that MV
plications or death. infection starts by infection of CD150+ alveolar macro-
phages or the binding to DC-SIGN+ submucosal DCs in
The current knowledge regarding the pathogenesis of the lumen of the respiratory tract (Figure 1a,b) [26–30].
measles thus owes much to in vivo studies in non-human An alternative route of entry is via the conjunctiva,
primates (NHPs), which are naturally susceptible to MV which contains susceptible CD150+ myeloid and lym-
infection. They are the ideal animal model to study measles phoid cells. Eye protection during contact with measles
pathogenesis. MV infection of New World monkeys can patients reduces the risk of MV infection, suggesting
lead to severe or even lethal outcomes, deeming them that, although to a lesser degree, MV can use the ocular
unsuitable as animal model [19,20]. However, in a recent route to enter the host [31]. Simultaneous intranasal,
study squirrel monkeys (Saimiri sciureus), a New World intratracheal and ocular administration of rCDV expres-
species, closely reproduced clinical symptoms of measles sing different fluorescent reporter proteins to ferrets
in humans [21]. Unfortunately, this model is still limited by resulted in multicolour viremia, showing that morbilli-
the lack of cross-reactive reagents. Rhesus (Macaca mulatta) viruses are able to enter the host at different anatomical
and cynomolgus macaques (Macaca fascicularis) present locations, as long as susceptible myeloid or lymphoid
similar clinical symptoms and virological, immunological cells are accessible [32].

infection systemically either through the circulatory system (e) or the lymphatic system (f). Infection of respiratory epithelial cells from the
basolateral side, combined with epithelial damage, leads to the release of infectious MV particles into the lumen of the respiratory tract
(g). Infection of dermal myeloid and lymphoid cells is followed by the spread of the infection among epidermal keratinocytes in a nectin-4-
dependent manner (h). MV-specific lymphocytes start to expand to clear the infection (i). Hyperaemia of the skin capillaries leads to the
recruitment of MV-specific lymphocytes and activated macrophages to infected skin areas and, together with oedema, gives the appearance of
erythematous, morbilliform skin rash (j). Upon recovery, loss of lymphocytes is partly masked by the expansion of newly generated cells (k). Skin
rash disappears and the epidermis is cleared of infection (l). The patient is protected against clinical measles for life. However, due to the loss of
pre-existing lymphocytes and plasma cells (m) and, consequently, pre-existing antibodies (n), the patient becomes susceptible to other infectious
diseases, especially respiratory infectious diseases. (o) The schematic representation of lymphopenia and changes in the composition of
lymphocyte populations before and after measles. The lymphotropic nature of MV leads to the infection and depletion of CD150+ pre-existing
lymphocytes, but the long-term loss is masked by the rapid generation of MV-specific cells and the expansion of new cells due to homeostatic
responses. (p) The types of cells infected by MV over time. Myeloid cells act as initial target cells and ‘Trojan horses’ by transporting virus to
CD150+ cells in lymphoid tissues. Lymphocytes play a major role in systemic dissemination, while respiratory epithelial cells are crucial in viral
transmission. The dermal myeloid and lymphoid cells and the epidermal keratinocytes are responsible for the appearance of skin rash. MV-
infected cells are mostly cleared by MV-specific immune cells but some lymphocytes still harbour MV RNA long after recovery from measles.

www.sciencedirect.com Current Opinion in Virology 2020, 41:31–37


34 Measles

Local and systemic dissemination of MV cells enter the dermis through the circulation. Here they
MV infection spreads as infected cells migrate to bron- interact with skin-resident myeloid and lymphoid cells in
chus-associated lymphoid tissues or draining lymph nodes the dermal papillae or near hair follicles or sebaceous
(Figure 1c,d) [29,33]. In addition to migrating MV- glands, where blood vessels and capillaries are abundantly
infected cells, MV can be carried by DC-SIGN+ DCs present (Figure 1h) [26,40,41]. Infection in the dermis is
and use these cells as a ‘Trojan horse’ to disseminate the disseminated to the keratinocytes in the epidermis and
virus systemically. Through cell-to-cell interaction, MV the virus spreads apically and laterally in a nectin-4-
spreads to susceptible CD150+ lymphocytes in the tissue- dependent manner [42]. In experimentally infected
associated lymphoid tissues. CD150 is abundantly NHPs, MV-infected cells were present in the dermis at
expressed in T cells, with higher expression in memory the peak of viremia (9 days post-inoculation) and the
cells than in the naive cells. In B cells, the dichotomy is infection continued to spread to the epidermis the fol-
less prominent, since both naive and memory cells lowing days [42]. At this time point, MV-specific IgM
express high levels of CD150. As a consequence, memory antibodies can be detected, demonstrating expansion of
T cells and naive and memory B cells are predominant MV-specific lymphocytes (Figure 1i) [43]. Maculopapular
target cells for MV infection in the lymphoid tissues skin rash appears around 11 days post-inoculation, during
[34,35,36]. The large number and the close interaction which inflammatory responses and mononuclear cell infil-
of susceptible myeloid and lymphoid cells in the draining tration are detected in the dermis and epidermis
lymph nodes allow rapid dissemination throughout the (Figure 1j) [42]. The appearance of the erythematous,
tissues [29]. Newly MV-infected lymphocytes subse- morbilliform skin rash possibly originates from the
quently leave the tissues and disseminate systemically, appearance of oedema and hyperaemia as a response to
leading to a cell-associated viremia (Figure 1e,f), and MV infection of the skin [42]. The importance of the host
spread the infection to other tissues and organs, such immune response in the pathogenesis of measles skin
as peripheral lymph nodes, thymus, bone marrow, spleen, rash is demonstrated by the absence of skin rash in
gastrointestinal tract, kidney, liver and skin. The mobility immunocompromised patients [44].
of MV-infected cells, the accessibility of organs to those
cells and the availability of susceptible cells in different Recovery phase of measles
organs are important factors during the systemic dissemi- The majority of MV-infected cells either die due to
nation of MV. infection or are cleared by MV-specific CD8+ T cells
[45,46]. New lymphocytes, either MV-specific or freshly
Prodromal phase of measles
recruited from the bone marrow, soon become available in
During systemic dissemination, early acute measles
lymphoid tissues and peripheral blood and the number of
patients exhibit coughing and fever, sometimes accom-
lymphocytes recovers to normal level (Figure 1k). During
panied by diarrhoea and/or vomiting and conjunctivitis,
recovery, new epithelial cells and keratinocytes proliferate
and become lymphopenic. Patients are highly contagious
and differentiate to make up losses (Figure 1l). Although
during the prodrome, and infectious virus can be isolated
recovery is often without complications, rare but severe
from throat and nose swabs, with higher virus titres found
measles-associated central nervous system (CNS) compli-
in the nose than in the throat [36]. Respiratory epithelial
cations may occur: acute disseminated encephalomyelitis
cells become infected at this stage, through contact
(ADEM), measles inclusion body encephalitis (MIBE) or
between MV-infected lymphoid or myeloid cells in the
subacute sclerosing panencephalitis (SSPE). ADEM hap-
respiratory submucosa and nectin-4 on the basolateral
pens during the recovery phase and is immune-mediated,
side of these epithelial cells (Figure 1g). MV ribonucleo-
possibly caused by molecular mimicry based on similar
protein (RNP) genomic complexes can be trafficked
structure between MV proteins and myelin [47]. MIBE
laterally in the respiratory epithelium through F-actin
occurs in young infants or immunocompromised patients
rings, demonstrating direct cell-to-cell transmission of
after several days or months after acute measles or vacci-
MV [37,38]. MV infection of the epithelial cells subse-
nation [48]. SSPE, unlike other measles-related CNS
quently leads to the production of new virus particles at
complications, is exclusively caused by wild-type MV
the apical cell surface. The new virus particles are then
infection [49]. How MV spreads to and in the CNS
released into the mucus that lines the lumen of the
remains unknown, although recent studies have shown
respiratory tract. Simultaneously, epithelial damage in
that hyperfusogenic viral complexes can mediate CD150-
the bronchi and bronchioles induces coughing. Combined
independent and nectin-4-independent viral entry. This
with the lack of nectin-4 expression at the luminal surface
hyperfusogenicity is caused by amino acid substitutions in
of epithelial cells, this results in unimpeded expel of
the ectodomain of the MV fusion (F) glycoprotein, which
infectious virions in large droplets or small aerosols
are reported in SSPE and MIBE cases [50,51,52]. Another
(Figure 1g) [33,39].
recent study also describes the transfer of cytoplasmic
Symptomatic phase of measles cargo, including infectious materials, from cell to cell
The emergence of skin rash marks the beginning of through nectin-1-nectin-4 interaction. Consequently,
clinical measles. MV-infected myeloid and lymphoid nectin-4+ epithelial cells can transfer infectious MV

Current Opinion in Virology 2020, 41:31–37 www.sciencedirect.com


Measles pathogenesis, immune suppression and animal models Laksono et al. 35

RNP to nectin-1+ primary neurons [53]. To date, there is which are responsible for suppression of T cell responses
no treatment for measles-associated CNS complications after immune activation [36,60]. B cell receptor sequenc-
and hence MV infection in the CNS will ultimately result ing of naive B cells from the paired PBMC samples
in death. revealed incomplete recovery of the genetic composition
of naive B cells with signatures of immunological imma-
MV genomic RNA has been reported to persist in experi- turity, consistent with the observed influx of transitional B
mentally infected NHPs up to 50 days post-inoculation in cells in these paired samples [59]. The masking effect by
peripheral blood mononuclear cells (PBMC) and 71 days newly generated cells, such as transitional B cells and
post-inoculation in lymph nodes. The decline of the RNA regulatory T cells, complicates the observation of measles-
in blood occurred in three phases: a rapid decline due to associated immune suppression, since there is no clear
clearance of infected cells (10–14 days post-inoculation), discrimination between the direct effect of MV infection
a transient increase up to 10-fold (14–24 days post- on the lymphocyte population and the host responses to
inoculation) and a slow decrease to undetectable levels infection and lymphopenia (Figure 1o).
(24 days or more after inoculation). This indicates that MV
might persist in the host for much longer than generally Vaccination of NHPs with vaccine strain-based rMV
recognised [54,55]. Persistence of MV RNA has also been expressing a fluorescent reporter protein did not result
reported in naturally MV-infected Zambian children up to in systemic replication and viremia, and instead resulted in
four months after the onset of skin rash [56]. high levels of protection from wild-type MV infection [61].
Consistent with this finding, no overall change in antibody
repertoire was observed in measles-mumps-rubella vacci-
Measles-associated immune suppression nated children [58]. Altogether, these observations signify
Several mechanisms have been proposed to explain mea- that measles vaccination does not lead to immune amne-
sles-associated immune suppression, such as suppression sia, but rather protects the host from it [62].
of lymphocyte proliferation, altered cytokine profiles,
apoptosis of bystander lymphocytes, and infection and
depletion of pre-existing CD150+ DCs and lymphocytes Conclusions
[57]. On the basis of the preferential MV infection of Experimental MV infections in primate models alongside
memory lymphocytes in experimentally infected NHPs, clinical studies in unvaccinated measles patients have
we proposed immune amnesia as a mechanism of measles- shed light on the complicated nature of MV as a
associated immune suppression [34]. This tropism was dual-tropic virus that can infect different types of cells
confirmed in PBMC collected from early acute measles (Figure 1p). Moreover, it was elucidated how MV
patients. Moreover, in paired PBMC samples collected lymphotropism drives measles-associated immune sup-
from unvaccinated children before and after measles there pression. Understanding the tropism and pathogenesis of
was a significant reduction in the percentages of circulat- MV in turn underlines the fact that measles is not an
ing memory T and B cell subsets [36]. This reduction was innocent childhood disease and that measles vaccination
detectable more than a month after recovery. Simulta- not only protects against measles, but indirectly also
neously, MV infection led to the loss of pre-existing against other infectious diseases.
antibodies in the plasma isolated from the same paired
samples, most likely due to MV infection and depletion of Conflict of interest statement
plasma cells in the bone marrow [58]. Accordingly, B cell Nothing declared.
receptor sequencing of the memory B cell pool from the
paired PBMC samples showed reduction of pre-existing B
References and recommended reading
cell memory clones, which were not reconstituted after Papers of particular interest, published within the period of review,
measles [59]. The combined loss of memory T and B cells have been highlighted as
and, ultimately, pre-existing antibodies through the loss  of special interest
of long-lived plasma cells, causes immune amnesia
(Figure 1m,n). Children are more likely to suffer from 1. Rima B, Balkema-Buschmann A, Dundon WG, Duprex P,
non-measles infectious diseases, especially upper respira- Easton A, Fouchier R, Kurath G, Lamb R, Lee B, Rota P et al.: ICTV
virus taxonomy profile: paramyxoviridae. J Gen Virol 2019,
tory infections, which may last several years after recovery 100:1593-1594.
from measles [2,3]. 2. Mina MJ, Metcalf CJ, de Swart RL, Osterhaus AD, Grenfell BT:
Long-term measles-induced immunomodulation increases
overall childhood infectious disease mortality. Science 2015,
The significant reductions in the frequency of some 348:694-699.
circulating lymphocyte subsets after measles is accompa-
3. Gadroen K, Dodd CN, Masclee GMC, de Ridder MAJ, Weibel D,
nied by increased frequencies of others. The most inter- Mina MJ, Grenfell BT, Sturkenboom M, van de Vijver D, de
esting subsets that increase in relative size after measles Swart RL: Impact and longevity of measles-associated
immune suppression: a matched cohort study using data from
are transitional B cells, which are bone marrow emigrants the THIN general practice database in the UK. BMJ Open 2018,
with reduced proliferation capacity, and regulatory T cells, 8:e021465.

www.sciencedirect.com Current Opinion in Virology 2020, 41:31–37


36 Measles

4. Patel MK, Dumolard L, Nedelec Y, Sodha SV, Steulet C, Gacic- macaques (Macaca mulatta) with different wild-type measles
Dobo M, Kretsinger K, McFarland J, Rota PA, Goodson JL: viruses. J Gen Virol 2007, 88:2028-2034.
Progress toward regional measles elimination - worldwide,
2000-2018. MMWR Morb Mortal Wkly Rep 2019, 68:1105-1111. 23. McChesney MB, Miller CJ, Rota PA, Zhu YD, Antipa L,
Lerche NW, Ahmed R, Bellini WJ: Experimental measles. I.
5. Navaratnarajah CK, Generous AR, Yousaf I, Cattaneo R: Pathogenesis in the normal and the immunized host. Virology
Receptor-mediated cell entry of paramyxoviruses: 1997, 233:74-84.
mechanisms, and consequences for tropism and
pathogenesis. J Biol Chem 2020, 295:2771-2786. 24. Auwaerter PG, Rota PA, Elkins WR, Adams RJ, DeLozier T, Shi Y,
Bellini WJ, Murphy BR, Griffin DE: Measles virus infection in
6. Buckland R, Wild TF: Is CD46 the cellular receptor for measles rhesus macaques: altered immune responses and
virus? Virus Res 1997, 48:1-9. comparison of the virulence of six different virus strains. J
Infect Dis 1999, 180:950-958.
7. Tatsuo H, Ono N, Tanaka K, Yanagi Y: SLAM (CDw150) is a
 cellular receptor for measles virus. Nature 2000, 406:893-897. 25. Ludlow M, Rennick LJ, Sarlang S, Skibinski G, McQuaid S,
First description of CD150 as a cellular receptor of MV. Moore T, de Swart RL, Duprex WP: Wild-type measles virus
infection of primary epithelial cells occurs via the basolateral
8. Noyce RS, Bondre DG, Ha MN, Lin LT, Sisson G, Tsao MS, surface without syncytium formation or release of infectious
 Richardson CD: Tumor cell marker PVRL4 (nectin 4) is an virus. J Gen Virol 2010, 91:971-979.
epithelial cell receptor for measles virus. PLoS Pathog 2011, 7:
e1002240. 26. de Swart RL, Ludlow M, de Witte L, Yanagi Y, van Amerongen G,
First description of nectin-4 as a cellular receptor of MV. McQuaid S, Yuksel S, Geijtenbeek TB, Duprex WP, Osterhaus AD:
Predominant infection of CD150+ lymphocytes and dendritic
9. Muhlebach MD, Mateo M, Sinn PL, Prufer S, Uhlig KM, cells during measles virus infection of macaques. PLoS Pathog
Leonard VH, Navaratnarajah CK, Frenzke M, Wong XX, 2007, 3:e178.
Sawatsky B et al.: Adherens junction protein nectin-4 is the
epithelial receptor for measles virus. Nature 2011, 480:530-533. 27. de Vries RD, Lemon K, Ludlow M, McQuaid S, Yuksel S, van
Amerongen G, Rennick LJ, Rima BK, Osterhaus AD, de Swart RL
10. Abdullah H, Brankin B, Brady C, Cosby SL: Wild-type measles et al.: In vivo tropism of attenuated and pathogenic measles
virus infection upregulates poliovirus receptor-related 4 and virus expressing green fluorescent protein in macaques. J Virol
causes apoptosis in brain endothelial cells by induction of 2010, 84:4714-4724.
tumor necrosis factor-related apoptosis-inducing ligand. J
Neuropathol Exp Neurol 2013, 72:681-696. 28. Mesman AW, de Vries RD, McQuaid S, Duprex WP, de Swart RL,
Geijtenbeek TB: A prominent role for DC-SIGN+ dendritic cells
11. Brancati F, Fortugno P, Bottillo I, Lopez M, Josselin E, Boudghene- in initiation and dissemination of measles virus infection in
Stambouli O, Agolini E, Bernardini L, Bellacchio E, Iannicelli M non-human primates. PLoS One 2012, 7:e49573.
et al.: Mutations in PVRL4, encoding cell adhesion molecule
nectin-4, cause ectodermal dysplasia-syndactyly syndrome. 29. Lemon K, de Vries RD, Mesman AW, McQuaid S, van
Am J Hum Genet 2010, 87:265-273. Amerongen G, Yuksel S, Ludlow M, Rennick LJ, Kuiken T, Rima BK
et al.: Early target cells of measles virus after aerosol infection
12. Dorig RE, Marcil A, Chopra A, Richardson CD: The human Cd46 of non-human primates. PLoS Pathog 2011, 7:e1001263.
molecule is a receptor for measles-virus (Edmonston Strain).
Cell 1993, 75:295-305. 30. Allen IV, McQuaid S, Penalva R, Ludlow M, Duprex WP, Rima BK:
Macrophages and dendritic cells are the predominant cells
13. Naniche D, Varior-Krishnan G, Cervoni F, Wild TF, Rossi B, infected in measles in humans. mSphere 2018, 3:e00570-
Rabourdin-Combe C, Gerlier D: Human membrane cofactor 00517.
protein (CD46) acts as a cellular receptor for measles virus. J
Virol 1993, 67:6025-6032. 31. Papp K: Experiments proving that the route of infection in
measles is the contamination of the conjunctival mucosa. Rev
14. de Witte L, de Vries RD, van der Vlist M, Yuksel S, Litjens M, de Immunol Ther Antimicrob 1956, 20:27-36.
Swart RL, Geijtenbeek TB: DC-SIGN and CD150 have distinct
roles in transmission of measles virus from dendritic cells to T- 32. de Vries RD, Ludlow M, de Jong A, Rennick LJ, Verburgh RJ, van
lymphocytes. PLoS Pathog 2008, 4:e1000049. Amerongen G, van Riel D, van Run P, Herfst S, Kuiken T et al.:
Delineating morbillivirus entry, dissemination and airborne
15. van der Vlist M, de Witte L, de Vries RD, Litjens M, de Jong MA,
transmission by studying in vivo competition of multicolor
Fluitsma D, de Swart RL, Geijtenbeek TB: Human Langerhans
canine distemper viruses in ferrets. PLoS Pathog 2017, 13:
cells capture measles virus through Langerin and present viral
e1006371.
antigens to CD4(+) T cells but are incapable of cross-
presentation. Eur J Immunol 2011, 41:2619-2631. 33. Ludlow M, McQuaid S, Milner D, de Swart RL, Duprex WP:
Pathological consequences of systemic measles virus
16. von Messling V, Springfeld C, Devaux P, Cattaneo R: A ferret
infection. J Pathol 2015, 235:253-265.
model of canine distemper virus virulence and
immunosuppression. J Virol 2003, 77:12579-12591. 34. de Vries RD, McQuaid S, van Amerongen G, Yuksel S,
Verburgh RJ, Osterhaus AD, Duprex WP, de Swart RL: Measles
17. Nambulli S, Sharp CR, Acciardo AS, Drexler JF, Duprex WP:
Mapping the evolutionary trajectories of morbilliviruses: what, immune suppression: lessons from the macaque model. PLoS
Pathog 2012, 8:e1002885.
where and whither. Curr Opin Virol 2016, 16:95-105.
18. Enkirch T, von Messling V: Ferret models of viral pathogenesis. 35. Laksono BM, Grosserichter-Wagener C, de Vries RD,
Virology 2015, 479-480:259-270. Langeveld SAG, Brem MD, van Dongen JJM, Katsikis PD,
Koopmans MPG, van Zelm MC, de Swart RL: In vitro measles
19. Albrecht P, Lorenz D, Klutch MJ, Vickers JH, Ennis FA: Fatal virus infection of human lymphocyte subsets demonstrates
measles infection in marmosets pathogenesis and high susceptibility and permissiveness of both naive and
prophylaxis. Infect Immun 1980, 27:969-978. memory B cells. J Virol 2018, 92:e00131-00118.

20. Lorenz D, Albrecht P: Susceptibility of tamarins (Saguinus) to 36. Laksono BM, de Vries RD, Verburgh RJ, Visser EG, de Jong A,
measles virus. Lab Anim Sci 1980, 30:661-665.  Fraaij PLA, Ruijs WLM, Nieuwenhuijse DF, van den Ham HJ,
Koopmans MPG et al.: Studies into the mechanism of measles-
21. Delpeut S, Sawatsky B, Wong XX, Frenzke M, Cattaneo R, von associated immune suppression during a measles outbreak in
Messling V: Nectin-4 interactions govern measles virus the Netherlands. Nat Commun 2018, 9:4944.
virulence in a new model of pathogenesis, the squirrel monkey First demonstration of preferential MV infection of B cells and memory T
(Saimiri sciureus). J Virol 2017, 91:e02490-02416. cells in human PBMC.
22. El Mubarak HS, Yuksel S, van Amerongen G, Mulder PG, 37. Shirogane Y, Watanabe S, Yanagi Y: Cooperation between
Mukhtar MM, Osterhaus AD, de Swart RL: Infection of different variants: a unique potential for virus evolution. Virus
cynomolgus macaques (Macaca fascicularis) and rhesus Res 2019, 264:68-73.

Current Opinion in Virology 2020, 41:31–37 www.sciencedirect.com


Measles pathogenesis, immune suppression and animal models Laksono et al. 37

38. Cifuentes-Munoz N, Dutch RE, Cattaneo R: Direct cell-to-cell dependent on hemagglutinin and hyperfusogenic fusion
transmission of respiratory viruses: the fast lanes. PLoS protein. J Virol 2018, 92:e02166-02117.
Pathog 2018, 14:e1007015.
52. Watanabe S, Shirogane Y, Sato Y, Hashiguchi T, Yanagi Y: New
39. Frenzke M, Sawatsky B, Wong XX, Delpeut S, Mateo M,  insights into measles virus brain infections. Trends Microbiol
Cattaneo R, von Messling V: Nectin-4-dependent measles virus 2019, 27:164-175.
spread to the cynomolgus monkey tracheal epithelium: role of Good review of studies demonstrating the crucial role of MV hyperfuso-
infected immune cells infiltrating the lamina propria. J Virol genicity during viral spread within the brain.
2013, 87:2526-2534.
53. Generous AR, Harrison OJ, Troyanovsky RB, Mateo M,
40. Tirado M, Adamzik K, Boer-Auer A: Follicular necrotic  Navaratnarajah CK, Donohue RC, Pfaller CK, Alekhina O,
keratinocytes - a helpful clue to the diagnosis of measles. J Sergeeva AP, Indra I et al.: Trans-endocytosis elicited by
Cutan Pathol 2015, 42:632-638. nectins transfers cytoplasmic cargo, including infectious
material, between cells. J Cell Sci 2019, 132:jcs235507.
41. Liersch J, Omaj R, Schaller J: Histopathological and
Demonstration of trans-endocytosis as a potential mechanism of MV
immunohistochemical characteristics of measles exanthema:
entry into neuronal cells.
a study of a series of 13 adult cases and review of the
literature. Am J Dermatopathol 2019, 41:914-923. 54. Lin WH, Kouyos RD, Adams RJ, Grenfell BT, Griffin DE: Prolonged
42. Laksono BM, Fortugno P, Nijmeijer BM, de Vries RD, Cordisco S, persistence of measles virus RNA is characteristic of primary
van Nieuwkoop S, Kuiken T, Geijtenbeek TBH, Duprex WP, infection dynamics. Proc Natl Acad Sci U S A 2012, 109:14989-
Brancati F et al.: Measles skin rash: infection of lymphoid and 14994.
myeloid cells in the dermis precedes viral dissemination to
55. Lin WH, Pan CH, Adams RJ, Laube BL, Griffin DE: Vaccine-
keratinocytes in the epidermis. bioRxiv 2019:867531.
induced measles virus-specific T cells do not prevent infection
43. van Binnendijk RS, van der Heijden RW, van Amerongen G, or disease but facilitate subsequent clearance of viral RNA.
UytdeHaag FG, Osterhaus AD: Viral replication and development mBio 2014, 5:e01047.
of specific immunity in macaques after infection with different
measles virus strains. J Infect Dis 1994, 170:443-448. 56. Riddell MA, Moss WJ, Hauer D, Monze M, Griffin DE: Slow
clearance of measles virus RNA after acute infection. J Clin
44. de Swart RL, Wertheim-van Dillen PM, van Binnendijk RS, Virol 2007, 39:312-317.
Muller CP, Frenkel J, Osterhaus AD: Measles in a Dutch hospital
introduced by an immuno-compromised infant from Indonesia 57. de Vries RD, de Swart RL: Measles immune suppression:
infected with a new virus genotype. Lancet 2000, 355:201-202. functional impairment or numbers game? PLoS Pathog 2014,
10:e1004482.
45. de Vries RD, Yuksel S, Osterhaus AD, de Swart RL: Specific CD8
(+) T-lymphocytes control dissemination of measles virus. Eur 58. Mina MJ, Kula T, Leng Y, Li M, de Vries RD, Knip M, Siljander H,
J Immunol 2010, 40:388-395.  Rewers M, Choy DF, Wilson MS et al.: Measles virus infection
diminishes preexisting antibodies that offer protection from
46. Morris SE, Yates AJ, de Swart RL, de Vries RD, Mina MJ, other pathogens. Science 2019, 366:599-606.
Nelson AN, Lin WW, Kouyos RD, Griffin DE, Grenfell BT: Modeling Demonstration that measles results in partial elimination of pre-existing
the measles paradox reveals the importance of cellular antibodies against other infections.
immunity in regulating viral clearance. PLoS Pathog 2018, 14:
e1007493. 59. Petrova VN, Sawatsky B, Han AX, Laksono BM, Walz L, Parker E,
 Pieper K, Anderson CA, de Vries RD, Lanzavecchia A et al.:
47. Fujinami RS, Oldstone MB, Wroblewska Z, Frankel ME, Incomplete genetic reconstitution of B cell pools contributes
Koprowski H: Molecular mimicry in virus infection: to prolonged immunosuppression after measles. Sci Immunol
crossreaction of measles virus phosphoprotein or of herpes 2019, 4:eaay6125.
simplex virus protein with human intermediate filaments. Proc Demonstration of depletion of previously expanded B cell populations
Natl Acad Sci U S A 1983, 80:2346-2350. and incomplete reconstitution of the naı̈ve B cell pool, based on B cell
receptor sequencing.
48. Baldolli A, Dargere S, Cardineau E, Vabret A, Dina J, de La
Blanchardiere A, Verdon R: Measles inclusion-body 60. Yu XL, Cheng YM, Shi BS, Qian FX, Wang FB, Liu XN, Yang HY,
encephalitis (MIBE) in a immunocompromised patient. J Clin Xu QN, Qi TK, Zha LJ et al.: Measles virus infection in adults
Virol 2016, 81:43-46. induces production of IL-10 and is associated with increased
49. Garg RK, Mahadevan A, Malhotra HS, Rizvi I, Kumar N, Uniyal R: CD4+ CD25+ regulatory T cells. J Immunol 2008, 181:7356-
Subacute sclerosing panencephalitis. Rev Med Virol 2019, 29: 7366.
e2058.
61. de Swart RL, de Vries RD, Rennick LJ, van Amerongen G,
50. Mathieu C, Ferren M, Jurgens E, Dumont C, Rybkina K, Harder O, McQuaid S, Verburgh RJ, Yuksel S, de Jong A, Lemon K,
Stelitano D, Madeddu S, Sanna G, Schwartz D et al.: Measles Nguyen DT et al.: Needle-free delivery of measles virus vaccine
virus bearing measles inclusion body encephalitis-derived to the lower respiratory tract of non-human primates elicits
fusion protein is pathogenic after infection via the respiratory optimal immunity and protection. NPJ Vaccines 2017, 2:22.
route. J Virol 2019, 93:e01862-01818.
62. Mina MJ: Measles, immune suppression and vaccination:
51. Sato Y, Watanabe S, Fukuda Y, Hashiguchi T, Yanagi Y, Ohno S: direct and indirect nonspecific vaccine benefits. J Infect 2017,
Cell-to-cell measles virus spread between human neurons is 74(Suppl. 1):S10-S17.

www.sciencedirect.com Current Opinion in Virology 2020, 41:31–37

You might also like