Sei sulla pagina 1di 13

Journal of Pathology

J Pathol 2015; 235: 253–265 INVITED REVIEW


Published online in Wiley Online Library
(wileyonlinelibrary.com) DOI: 10.1002/path.4457

Pathological consequences of systemic measles virus infection


Martin Ludlow,1 Stephen McQuaid,2 Dan Milner,3,4 Rik L de Swart5 and W Paul Duprex1*
1 Department of Microbiology, Boston University School of Medicine, MA, USA
2 Tissue Pathology Laboratories, Belfast Health and Social Care Trust, Northern Ireland
3 Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, USA
4 Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
5 Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands

*Correspondence to: W Paul Duprex, Department of Microbiology, Boston University School of Medicine, 620 Albany Street, Boston, MA 02118,
USA. E-mail: pduprex@bu.edu

Abstract
The identification of poliovirus receptor-like 4 (PVRL4) as the second natural receptor for measles virus (MV)
has closed a major gap in our understanding of measles pathogenesis, and explains how this predominantly
lymphotropic virus breaks through epithelial barriers to transmit to a susceptible host. Advances in the development
of wild-type, recombinant MVs which express fluorescent proteins making infected cells readily detectable in living
tissues and animals, has also increased our understanding of this important and highly transmissible human disease.
Thus, it is timely to review how these advances have provided new insights into MV infection of immune, epithelial
and neural cells. This demands access to primate samples that help us understand the early and acute stages of
the disease, which are challenging to dissect due to the mild/self-limiting nature of the infection. It also requires
well-characterized and rather rare human tissue samples from patients who succumb to neurological sequelae to
help study the consequences of the long-term persistence of this RNA virus in vivo. Collectively, these studies
have provided unique insights into how the use of two cellular receptors, CD150 and PVRL4, governs the in vivo
tissue-specific temporal patterns of virus spread and resulting pathological lesions. Analysis of tissue samples has
also demonstrated the importance of differing mechanisms of virus cell-to-cell spread within lymphoid, epithelial
and neural tissues in the dissemination of MV during acute and long-term persistent infections. Given the incentive
to eradicate MV globally, and the inevitable question as to whether or not vaccination should cease in light of the
existence of closely related morbilliviruses, a thorough understanding of measles pathological lesions is essential.
Copyright © 2014 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.

Keywords: measles virus; cell-to-cell spread; immune system; epithelium; central nervous system

Received 22 August 2014; Revised 30 September 2014; Accepted 3 October 2014

No conflicts of interest were declared.

Introduction make it timely to update and consolidate the current


understanding of MV pathology.
It has been estimated that 7–8 million children died each The history of measles spans a millennium since the
year due to measles virus infections in the pre-vaccine prototypic morbillivirus MV was first described by Al
era. That there were 122 000 measles deaths globally Rhazes of Baghdad [3]. Exactly when the virus first
in 2012, and that vaccination resulted in a 78% drop entered a human population is debatable, and its origin is
between 2000 and 2012 worldwide, is a testament even less clear. However, it is certain that endemic trans-
to the perseverance and commitment of the World mission requires populations sizes of several hundred
Health Organization (WHO). Currently, WHO aims to thousand individuals [4], due to the basic reproductive
reduce the overall mortality rate in under-5 year-olds by number (R0 ) of the virus (∼15–17), which is one of the
two-thirds between 1990 and 2015 [1]. Over 1 billion highest for any human pathogen [5,6] and the fact that
children have been vaccinated against measles and MV infection leads to lifelong immunity [7]. This high
other childhood diseases since 2000. The Measles and level of transmissibility also demands that vaccination
Rubella (M&R) Initiative proposes, by 2015, to reduce rates of >95% are achieved to ensure herd immunity
measles related deaths by 95% compared to the year [8]. It is generally accepted that MV, like many other
2000 levels, and to eliminate the virus from at least five human pathogens, crossed the species barrier from an
designated WHO regions by 2020 [2]. These impressive animal reservoir. Rinderpest virus is closely related to
achievements in the public health arena, alongside a MV and has been suggested to be the progenitor of the
number of significant advances in our understanding of human infection, although this is purely speculative
MV pathogenesis at the molecular and cellular levels, [9,10]. Closely related morbillivirus sequences have
Copyright © 2014 Pathological Society of Great Britain and Ireland. J Pathol 2015; 235: 253–265
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
254 M Ludlow et al

been found in vampire bats in South America [11] rare, meaning that much of what we know about the
and elsewhere [12], suggesting other possible origins. pathological lesions in prodromal and early acute stages
Although the evolutionary origins of MV are uncertain, comes from non-human primate infections [20–22]. A
this virus has evolved to become a pathogen of primates, major advance in understanding the early stages of the
with humans being the only species present in sufficient disease was achieved when reverse-genetics systems
numbers to maintain endemic transmission [4,13]. were developed for clinical isolates, which allowed the
In 2010 the WHO ad hoc Global Measles Advisory generation of recombinant MVs expressing enhanced
Group determined that it is possible to eradicate MV, green fluorescent protein (EGFP), which, in turn, per-
and a timeline will be set in 2015 [2]. To achieve this, mitted the highly sensitive detection of infected cells
it will be critical to attain and sustain high levels of in vivo [23,24]. Cynomolgus macaques are the optimal
immunity using a two-dose vaccination schedule. This animal model for measles virus pathogenesis, as they
is a challenge in both the developed and the develop- recapitulate many aspects of the disease in humans,
ing world, albeit for differing reasons. Logistics, vaccine can be naturally infected and recover fully from the
refrigeration, donor ‘fatigue’, ensuring a consistent sup- infection [20,25]. Combining wild-type recombinant
ply of vaccine at an appropriate cost, and the fact that (r) viruses, such as rMVIC323 EGFP (derived from a
a single dose of MV vaccine still leaves some child- Japanese wild-type MV) or rMVKS EGFP (derived from
ren unprotected, are some of the major challenges in a wild-type MV from Khartoum, Sudan), with this
the developing world. Complacency, unfounded links macaque model has been instrumental in helping to
between MV vaccination and a disparate array of uncon- shed new light on MV pathogenesis and transmission
nected conditions and diseases [14], lack of education [23,26–28]. This is in direct contrast to what we learn
about the seriousness of the disease, and a degree of from human tissue samples, since the majority of these
mistrust in the medical establishment have had a signif- are obtained post mortem, from individuals who have
icant impact on vaccine uptake in Europe and the USA, either succumbed to other infections or have died
where the target of cessation of endemic MV transmis- due to CNS complications, such as MIBE or SSPE
sion was attained in 2000 [15]. Consequently, measles (Figure 1A). Secondary infections are prevalent during
cases in the USA have reached a 20 year high in 2014, measles because of the profound immune suppres-
with 593 infections in 18 outbreaks representing nearly sion which accompanies the disease (Figure 1A). For
90% of cases; this is the highest number since MV example, bacterial pneumonitis and bronchiolitis are
was eliminated in 2000 [16]. During the most recent frequently observed complications (see below). These,
12 month reporting period, 30 European countries per- in addition to underlying infections, such as HIV-1,
forming measles surveillance recorded nearly 10 000 or malnutrition, make describing the pathological
cases, with Germany, Italy, The Netherlands, Romania consequences of the infection challenging.
and the UK accounting for just over 90% of these [17]. Our previous reviews have been virocentric, concen-
Unsurprisingly, the majority of affected individuals were trating on the molecular biology of MV and mutations in
unvaccinated, which remains a major challenge for pub- the genome that occur when the virus reactivates months
lic health and policy makers alike, given the drive to to years after the primary infection [14,29,30]. In this
eradicate the virus. review we focus on virus cell entry, since dissecting
this step has been critical in understanding acute viral
pathogenesis and MV transmission. Given that measles
is predominantly a disease of the immune system, this
Animal models and human tissues: pragmatic is where we start (Figures 1A, 1B, and 2). Recognizing
pathology that as a respiratory pathogen the virus must be expelled
from the respiratory tract to ensure transmission, we
Although measles is primarily a human disease, subsequently focus on how it crosses epithelial barriers
non-human primates are also susceptible to natural (Figures 1A, 1C, and 3). Finally, we address entry into,
MV infection [18,19]. Clinically, cough, conjunctivitis and spread within, the CNS, the one aspect of pathogen-
and coryza, colloquially the ‘three Cs’, are observed esis which remains enigmatic (Figures 1A, 1D, and 4).
at the end of the prodromal phase in humans, and
these mild symptoms precede the hallmark Koplik
spots in the cheeks, and rash which spreads from the Entry and spread: systemic lymphotropism
face to the extremities over subsequent days. Pyrexia
is also a common early sign of the infection and The lymphotropic nature of MV is well recognized as
lasts approximately 1 week. The disease course is a distinguishing feature of the disease, with giant cell
usually self-limiting and resolves after 3 weeks. As formation reported in tonsils, appendix, lymph nodes,
such, there is very limited clinical material available thymus and spleen [31–34]. Understanding how MV
from human cases in the prodromal [∼10–14 days spreads with such specific cellular tropism through the
post-infection (d.p.i.), early or established acute phases immune system is vital in pathogenesis studies. Identi-
(∼14–19 d.p.i.) of the disease (Figure 1A). Samples are fication of the cell surface receptor CD150 (SLAMF1),
sometimes available from vaccinated individuals with which is expressed on activated immune cells, pro-
undiagnosed immunodeficiencies but these are very vided the key starting point [35]. The molecule is a
Copyright © 2014 Pathological Society of Great Britain and Ireland. J Pathol 2015; 235: 253–265
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
Measles virus pathology 255

A progression

transmission

prodromal early acute established acute late

conjunctivitis ADME SSPE


coryza MIBE
Koplik’s spots

cough

rash

fever

secondary infections

immune suppression

immune
B

DCs macrophages B-cells T-cells

CD150 (activated immune cells)

C epithelia

simple columnar stratified squamous


PVRL4 (adherens junction)
D
neural
astrocvtes

neurons
oligodendrocytes

entry into CNS (?) and spread within CNS (?)

Figure 1. Summary of the clinical and pathological features of measles. (A) Temporal progression of measles clinical signs and disease
course. (B) Cellular tropism of MV in vivo is largely determined by the expression of the cellular receptors CD150 on subsets of immune
cells and PVRL4 on epithelial cells. The mechanism(s) of MV entry and spread in the CNS are undetermined.

member of the immunoglobulin (Ig) superfamily and is MV, it does not facilitate cell entry by mediating
expressed on subsets of B and T cells, dendritic cells, virus-to-cell fusion at the plasma membrane, the normal
macrophages, platelets and haematopoietic stem cells mode of morbillivirus entry into cells [39].
[36,37]. It functions as a co-stimulatory molecule and Early stages of infection have been examined
is involved in the regulation of innate and adaptive using viruses which express fluorescent proteins in
immune responses [36,38]. The role of the C type lectin both macaque and transgenic mouse models [23,40].
DC-SIGN in measles pathogenesis is still uncertain. Analysis of respiratory tract tissue from MV-infected
Although this molecule is able to bind to and capture macaques shows that MV-infected dendritic cells
Copyright © 2014 Pathological Society of Great Britain and Ireland. J Pathol 2015; 235: 253–265
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
256 M Ludlow et al

A DAPI/EGFP B

DAPI/CD11c/MV N

C DAPI/CYK/EGFP D EGFP E PI/EGFP

F EGFP G PI/EGFP H PI/EGFP

I DAPI/CYK/EGFP J DAPI/CD20/EGFP

Figure 2. MV-infection of the immune system of the macaque. MV-infected cells were detected directly in thin sections (A) and 200 μm
vibratome-cut sections (D–H) or indirectly by immunocytochemical (B inset, C, I, J) and immunohistochemical (B) staining. (A) Infection of
early target cells with the morphology of myeloid cells in the alveolar space; the walls of the alveoli are indicated with a dashed line. (B)
MV-infected cells in bronchial associated lymphoid tissue; (inset) MV-infected (green) CD11c+ myeloid cells (red) in the alveolar lumen of the
lung. (C) Syncytia (green) present in sub-epithelial lymphoid cells of the primary bronchus. (D) A syncytium of interconnected MV-infected
cells (green) in the tracheobronchial lymph node. (E) Individual and connected MV-infected immune cells in the tracheobronchial lymph
node. (F) Multiple MV-infected (green) B cell follicles in the spleen. (G) Higher-magnification image of B cell follicles in the spleen,
demonstrating specific targeting of these structures. (H) Dispersed MV-infected cells (green) in the spleen; (inset) at higher magnification,
connected cells are visible. (I) Aggregation of MV-infected immune cells (green) present beneath and within cytokeratin+ squamous
epithelium (red) of the tongue. (J) Infiltration of MV-infected (green) CD20+ B cells (red) into bronchial epithelium; the approximate
position of the bronchial lumen is indicated with a dashed line. Propidium iodide (red) was used as nuclear counterstain in plates (E, G, H);
DAPI was used in plates (A, B inset, C, I, J); square, B cells; triangle, T cells; diamond, dendritic cells; inverted triangle, macrophages.

and/or alveolar macrophages can be detected in the readily detected at the peak of infection (7–9 d.p.i.) in
lung as early as 2 (d.p.i.) (Figure 2A). Spread of bronchial lymphoid tissue (Figure 2C), and clusters of
virus to bronchus associated lymphoid tissue (BALT) MV-infected interconnected immune cells are present in
(Figure 2B) and aggregates of CD11c+ myeloid cells the trachea-bronchial lymph node (Figure 2D, E). Anal-
in alveolar spaces (Figure 2B, inset) results in local ysis of tissue sections from the spleen has shown that B
amplification of virus infection between 3–5 d.p.i. cell follicles are targets of MV infection (Figure 2F–G).
Following spread of the virus, giant cell formation is Scattered infected cells are also observed between
Copyright © 2014 Pathological Society of Great Britain and Ireland. J Pathol 2015; 235: 253–265
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
Measles virus pathology 257

follicles (Figure 2H). Lymphoid depletion is observed and pathological lesions. Further research using in vivo
in these follicles following the peak of infection at 11 models of measles may help to elucidate answers to the
d.p.i. [28], a phenomenon which has also been noted following questions:
as a feature of measles in humans [31]. MV-infected
immune cells contact and/or infiltrate the epithelium at • Does MV binding to CD150 stimulate or block
the peak of infection [27,41], providing a mechanism CD150-mediated cell signalling pathways in immune
through which virus is seeded into both squamous cells?
(Figure 2I) and columnar epithelia of the respiratory • Does MV vaccination provide lifelong immunity in
tract (Figure 2J). the absence of endemic virus transmission?
Morbilliviruses cause variable levels of immune • Should vaccination cease if it is possible to eradicate
suppression, leading to increased susceptibility to MV?
opportunistic infections [21,42], so that patients often • Can a better understanding of the lifelong immunity
develop potentially life-threatening complications, such afforded by MV infection lead to more effective,
as pneumonia or gastroenteritis [43–46]. Although rational vaccine design for other pathogens?
studied since the beginning of the twentieth century
[47], the relative importance of different mechanisms
leading to this immune suppression remains disputed. Exit and transmission: local epitheliotropism
Many studies have relied on in vitro observations,
making it very difficult to assess their importance for
immune suppression in vivo. Paradoxically, measles Textbooks traditionally suggested that epithelial cells
is also associated with immune activation [48,49]. were the primary target for MV. However, as outlined
Patients develop robust MV-specific humoral and cel- above, initial infection of immune system cells is essen-
lular immune responses that mediate the clearance of tial for MV entry and systemic cell-to-cell spread. Given
infected cells. Acute measles is associated with lym- that MV is a respiratory pathogen, this appears to be a
phopaenia, but peripheral blood lymphocyte numbers ‘suicidal strategy’. The discovery of PVRL4 (nectin 4)
normalize within days after resolution of the disease, as the second natural MV receptor shows how elegantly
whereas immune suppression extends over a prolonged the virus has evolved to cross epithelial barriers and
period of time. This rapid recovery has been used to ensure transmission to a susceptible host [54,55]. The
argue against lymphocyte depletion as a direct cause molecule is a component of adherens junctions, located
of measles immune suppression. Instead, research has at the basolateral side of epithelia, and is a member of
mainly focused on MV-mediated functional impair- the nectin family of adhesion molecules belonging to
ment of lymphocytes and/or antigen-presenting cells the Ig superfamily [56]. Subsequent studies demonstrat-
[50–52]. Following experimental MV infection of ing usage of canine PVRL4 by canine distemper virus
non-human primates, the virus predominantly replicates (CDV) and ovine PVRL4 by peste des petits ruminant
in memory T lymphocytes and follicular B lymphocytes virus confirmed that this molecule, like CD150, is also a
[28,53] (Figure 1B). Thus, infection and subsequent pan-morbillivirus receptor [57,58].
depletion of CD150+ antigen-experienced lymphocyte Focal areas of MV infection are present in multiple
subsets may be considered as a potential mediator epithelial tissues of the respiratory tract of MV-infected
of measles immune suppression [28]. Disappearance macaques, including pseudostratified columnar epithe-
of pre-existing memory lymphocytes is masked by lial cells in the trachea (Figure 3A) and non-keratinized
the rapid proliferation of newly induced MV-specific stratified squamous epithelial cells in the tongue
lymphocytes, thus explaining the measles paradox. (Figure 3B). We have also previously documented
Although infection of antigen-presenting cell popula- infection of simple squamous epithelial cells in alveoli
tions and epithelial damage may contribute to the risk and simple columnar epithelial cells in bronchi [26,53].
of bacterial infection or dissemination during the acute While much attention has been focused on MV-infected
phase of the disease, long-term immune suppression cells in the trachea [41,55], it is clear that multiple
mainly results from depletion of pre-existing memory subtypes of epithelial cells are susceptible to MV infec-
lymphocyte populations [125]. tion, with different epithelia displaying various levels
of pathological lesions [26,27]. This is most evident in
the upper respiratory tract, where very large numbers
of contiguous epithelial cells in the nasal cavity are
MV lymphotropism: future perspectives infected [26–28]. Macroscopic screening shows these
are primarily in ciliated regions of the nasal mucosa
Although immune suppression and lifelong immunity and that infection of the conchae is also pronounced.
afforded by primary MV infection are both defining These levels of infection probably explain the highly
features of measles, specific mechanisms leading to transmissible nature of MV, and it is reasonable to
immune system dysregulation remain controversial, suggest that virus shedding from here, rather than
and our understanding of why MV stimulates such an from the trachea, is important for host-to-host spread.
effective, long-lasting, immune response is still not Syncytia of fused MV-infected cells are more readily
well integrated with our knowledge of virus tropism detected in columnar than in squamous epithelium, with
Copyright © 2014 Pathological Society of Great Britain and Ireland. J Pathol 2015; 235: 253–265
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
258 M Ludlow et al

A EGFP B

D DAPI/CYK DAPI/MV N DAPI/CYK/MV N

E F DAPI/EGFP DAPI/EGFP

G DAPI/CD11c/MV N H DAPI/CYK/MV N

Figure 3. MV infection of macaque epithelial tissues. (A) MV-infected pseudostratified columnar epithelial cells in a 200 μm vibratome-cut
section of tracheal epithelium. (B–I) Immunocytochemically stained, microtome-cut epithelium tissue sections. (B) MV-infected stratified
squamous epithelial cells in the tongue. (C) Serial sections of primary bronchus; multinucleated giant cells (arrows) present in the epithelium
of an H&E-stained section (left) are positive for EGFP in a serial immunohistochemically stained section (right). (D) A large syncytium of
MV-infected cells in cytokeratin+ (CYK) pseudostratified columnar epithelial cells in the adenoid; single stains (left and middle panels), dual
label (right panel). (E) Exfoliation of a focus of MV-infected tracheal epithelial cells. (F) MV-infected epithelial cells (green) in the primary
bronchus undergoing exfoliation into the lumen; (right panel) higher magnification image of the boxed area. (G) CD11c+ myeloid cells (red)
are present beneath and within (arrows) MV-infected (green) bronchial epithelium. (H) Extensive disruption and dissolution of adenoidal
epithelium due to infiltration of MV-infected immune cells (green; arrows); MV-infected cytokeratin+ pseudostratified columnar epithelial
cells (green and red) are indicated by arrowheads. Propidium iodide (red) was used as nuclear counterstain in (A); DAPI in (D, F–H); triangle,
columnar; diamond, stratified squamous.

syncytia containing >30 nuclei readily detected in both pathological response may serve as one mechanism
bronchial and adenoidal epithelium (Figure 3C, D). through which virus is released into the environment
Exfoliation of MV-infected columnar epithelial cells following the induction of a cough response. We have
into the lumen of the respiratory tract is also observed in also observed extensive pathological lesions in the
both the trachea and the bronchus (Figure 3E, F). This respiratory epithelium in the latter stages of the disease
Copyright © 2014 Pathological Society of Great Britain and Ireland. J Pathol 2015; 235: 253–265
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
Measles virus pathology 259

(9–11 d.p.i.), with dissolution of epithelium observed well-delineated and distinct neurological complications
in the bronchus (Figure 3G) and more extensively have been described (reviewed in [72,73]).
in lymphoid tissues, such as adenoidal (Figure 3H) Acute demyelinating encephalomyelitis (ADME) is
or tonsillar tissue [26]. Such MV-infected epithelia a neurological complication which typically occurs in
are extensively infiltrated by immune cells, resulting children or young adults following a viral or bacte-
in disruption of epithelial integrity, which may con- rial infection [74]. A number of pathogens have been
tribute to the cell-free and cell-associated virus we have associated with ADME, including Epstein-Barr virus,
detected in throat brushings and nasal swabs from these herpes simplex virus, influenza virus and Mycoplasma
animals [26]. pneumonia [75,76]. A diagnosis of ADME has also
How the interaction of the MV H glycoprotein with been reported in approximately 1 of 1000 measles
PVRL4 might perturb the normal functioning of this cases following the systemic phase of the disease
molecule in adult epithelial tissues remains a topic for [77]. The pathogenesis of ADME is poorly understood
future investigation. PVRL4 has been shown to interact but is thought to have an autoimmune component,
with both PVRL1 via its extracellular V domain, and as myelin basic protein (MBP) can be detected in
with afadin via its cytoplasmic domain sequence [56]. cerebrospinal fluid in ADME patients, together with
Afadin is an F-actin-associated molecule that connects lymphocyte-proliferative responses against MBP [78].
the actin cytoskeleton to the nectin complex at the Very few studies have reported direct evidence of MV
adherens junction [59,60]. Although fetal tissue and infection, such as viral RNA or antigen in CSF or brain
adult placental tissue show high levels of PVRL4, only biopsies from ADME patients [79]. While many aspects
low levels of expression are found in other adult tissues, of ADME pathogenesis are unknown, perivenous infil-
including the trachea, lung and stomach [56,61,62]. tration with perivascular demyelination and associated
There have also been a number of reports investigat- white matter lesions have been described [77].
ing the efficacy of PVRL4 as a diagnostic marker Measles inclusion body encephalitis (MIBE) typ-
for breast, lung and ovarian cancer [62–65] and a ically occurs in immunocompromised individuals
recent study has shown that up-regulation of PVRL4 within 1 year after resolution of uncomplicated measles
promotes anchorage-independent growth of tumour [80,81]. The condition has been reported in children
cells [66]. with leukaemia or transplant patients undergoing an
immunosuppressive therapy regime and in children
co-infected with human immunodeficiency disease
MV epitheliotropism: future perspectives virus (HIV)-1 [82–84]. Few detailed pathological
descriptions of MIBE have been reported but infection
of neurons and glial cells has been observed in brain
While much progress has been made in understanding tissue from MIBE patients [85,86].
how the MV H glycoprotein interacts with PVRL4 from The term ‘subacute sclerosing panencephalitis’
a molecular perspective [67,68], many aspects of the (SSPE) was first used in 1950 to describe a ‘sporad-
pathological consequences of this interaction in vivo ically occurring European encephalitis’ [87]. It is a
with respect to MV cell-to-cell spread and transmission disease that develops in children 5–10 years after a nor-
remain to be determined, including: mal episode of natural measles, despite the presence of
MV-specific antibody titres [88], and is a paradigm for
• What is the distribution, spatial organization and
the long-term persistence of an RNA virus in humans.
expression level of PVRL4 in human tissues?
In rare cases, prolonged persistence of MV may lead
• Is it possible for MV to disrupt the integrity of the
to the development of SSPE in immunocompetent
epithelium by interacting with PVRL4 without infect-
adults, including a 49 year-old man who had measles
ing epithelial cells?
at the age of 2 [89]. Clinically, this inevitably fatal
• Do innate immune responses in infected epithelial
disease is characterized initially by subtle behavioural
cells contribute to the pathological lesions observed
changes, which lead progressively over a period of
within epithelia?
1–3 years to myoclonic jerks, ataxia and death, which
in many cases is due to pneumonia [90–92]. In some
cases a more rapid fulminant disease course has been
Acute and persistent: extreme neurotropism observed, particularly in adults [93,94]. The frequency
of SSPE has been re-evaluated in recent years, as the
Most cases of measles resolve without neurological reduction in overall numbers of measles cases following
sequelae following systemic virus spread. However, the introduction of an efficacious vaccine has made
studies of brain tissue obtained from autopsy sub- it easier to compare the number of SSPE cases to the
jects report the presence of MV mRNA in approxi- ‘true’ number of measles infections [95]. SSPE cases
mately 20% [69,70] and transient electroencephalogram have been reported at a rate of approximately 1:10
changes have been observed in children with uncompli- 000 natural measles infections in some subdistricts
cated measles [71]. It remains to be determined whether of Papua New Guinea [96,97], 1:4635 in the USA
these observations support the persistence of MV within [95] and as high as 1:1700–1:3300 for children in
the CNS in the absence of clinical disease, but three Germany [98].
Copyright © 2014 Pathological Society of Great Britain and Ireland. J Pathol 2015; 235: 253–265
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
260 M Ludlow et al

A viral aetiology for SSPE was first suggested follow- formation suggests that a novel mechanism of viral
ing the observation of intracytoplasmic and intranuclear transmission may mediate the spread of MV from
inclusion bodies in two cases of what was then termed neuron-to-neuron. We have also observed numerous
‘lethargic or epidemic encephalitis’ [99,100]. Later, MV-infected neurons with extended dendritic processes
viral particles characterized as paramyxovirus in origin in immunostained brain sections from the temporal lobe
were detected by electron microscopy in brain biopsies of an SSPE case (Figure 4A, 4B) and the frontal cortex
taken from SSPE patients [101]. Direct confirma- of an MIBE case (Figure 4D, 4E). No co-localization
tion that MV was pathologically linked to SSPE was was observed in the grey matter of an SSPE patient
obtained using MV-specific antisera to detect infected between MV antigen present in neuronal tracts and
neurons and glial cells in brain sections [88]. The NOGO, a marker for oligodendrocytes (Figure 4C).
pathology of SSPE is determined by a complex mixture While cell-to-cell transmission of nucleocapsids was
of the unique neuronal environment, host genetics and first suggested as a possible mechanism for the spread of
the trans-synaptic spread of mutant variants of MV. MV in the brains of SSPE patients 40 years ago [115],
Direct sequencing of MV from SSPE brain tissue has the specific mechanism(s) responsible for mediating
shown that viruses contain a multitude of mutations, spread across the synapse is still unknown. However,
leading to an absence of M protein in infected cells the recent report of a reverse-genetics system for an
and expression of F glycoproteins with truncated cyto- ‘SSPE virus’ could re-energize studies into neurological
plasmic tails [102–105]. For further information on the aspects of MV infections [116].
genetic variation displayed by ‘SSPE viruses’, we refer
readers to previous reviews we have published on this
subject [14,29]. MV infection of glial cells in the human CNS
Pathologically, the brains of SSPE patients are char-
acterized by inflammatory infiltrates in both the grey Although the presence of MV antigens in oligodendro-
and white matter, explaining the term ‘panencephalitis’. cytes is a characteristic feature of SSPE, the extent of
Diffuse demyelination, astroglial sclerosis and viral astrocyte cell infection in SSPE remains controversial,
antigen-containing inclusion bodies in neurons and as previous studies have reported the presence of MV N
oligodendrocytes are also observed [106]. While the protein in varying numbers of astrocytes [113,117,118].
neurological aspects of SSPE are well characterized, it We have observed abundant MV-positive cells with
is still unknown where MV ‘persists’ in the body prior the morphological appearance of oligodendrocytes, but
to the onset of overt clinical signs. Reports of MV RNA only limited numbers of infected astrocytes in the white
and/or antigen in peripheral lymphoid tissues of SSPE matter of SSPE cases (Figure 4F). Examination of white
patients are sparse [107–109] and thus must be treated matter areas on adjacent sections with triple-labelling
with caution. Similarly, the route of entry of MV into the immunofluorescence revealed abundant GFAP-positive
CNS is also unknown, but is assumed to occur via anal- astrocytes and CD68-positive foamy macrophages.
ogous mechanism(s) to CDV in members of the family However, MV-positive cells with the smaller, more
Canidae. Although MV infection has been reported in rounded morphology of oligodendrocytes were dis-
limited numbers of human cerebral endothelial cells and tinct from these two cell populations (Figure 4G–I).
larger numbers of cells in perivascular cuffs in SSPE Dual-labelling immunofluorescence staining with
brain tissue [110,111], studies have been limited by the anti-NOGO antibody confirmed that MV-infected
fact that biopsy and/or brains from SSPE patients are oligodendrocytes are also present in MIBE brain tissue
only available after the onset of neurological clinical (Figure 4J, 4K). It is assumed that virus spread in glial
signs. The role of CD150 and PVRL4 in mediating the cells occurs via interconnected cellular processes as
entry and/or cell-to-cell spread of MV within the CNS MV-induced fusion between neighbouring glial cells
is unknown. One study has reported CD150 expression has been observed in both a case of SSPE and a case of
in subsets of leukocytes in inflammatory infiltrates, but MIBE [119,120].
found that expression was absent in the rest of the brain
parenchyma [112].
MV neurotropism: future perspectives
MV infection of neurons in the human CNS Since MV was first isolated from brain biopsies obtained
from SSPE patients over 30 years ago, comparatively
Detailed pathological and ultrastructural studies of little progress has been made in delineating the patho-
SSPE brain tissue have indicated that the mechanism of genesis of this severe neurological disease. A number of
viral spread in the CNS is different from that observed questions remain to be elucidated:
in non-neuronal cells. Examination of brain tissue
from patients with MIBE and SSPE has shown that the • What is the specific mechanism(s) mediating
presence of MV antigen in neurons is a characteristic trans-synaptic MV spread?
feature [113,114]. The close proximity of MV-infected • How is the MV ribonucleoprotein transported along
interconnected neurons in the absence of syncytium axons?
Copyright © 2014 Pathological Society of Great Britain and Ireland. J Pathol 2015; 235: 253–265
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
Measles virus pathology 261

A B PI/MV N

C SSPE/NOGO/NFP D PI/MV N E PI/MV N

F PI/MV N G GFAP/MV N H GFAP/CD68/SSPE

I SSPE/CD68/GFAP J SSPE/NOGO/GFAP K SSPE/NOGO/GFAP

Figure 4. MV-infection of the human central nervous system. Detection of MV-infected cells in MIBE (A–C, I–K) and SSPE (D–H) human
brain tissue sections. MV antigen was detected by immunohistochemical (A) or immunocytochemical (B–K) staining. (A) Individual
MV-infected cortical neurons are observed in the grey matter. (B) Three MV-infected neurons in the grey matter are surrounded by multiple
infected neuronal processes. (C) MV antigen (blue) is associated with neuronal tracts (green) in the cortex, with no virus spread observed
to NOGO+ oligodendrocytes (red). (D) Detection of MV antigen (green) in neurons of the frontal cortex by tyramide signal amplification;
cytoplasmic inclusions of MV N protein are present in a neuronal cell body (arrow) and in a number of neuronal processes (arrowhead). (E)
A characteristic ‘beading’ pattern of aggregations of MV antigen is observed along dendritic processes of individual neurons. (F) Detection
of MV antigen (yellow) in oligodendrocytes in the white matter by tyramide signal amplification. (G) MV antigen (green) is restricted to
oligodendrocytes in the white matter; no dual labelling is observed with GFAP+ astrocytes (red) or CD68+ foamy macrophages (red). (H) No
virus spread is observed from MV-infected oligodendrocytes (green) in the white matter to adjacent to CD68+ foamy macrophages (red) or
GFAP+ astrocytes (blue). (I) MV-infection (blue) is restricted to oligodendrocytes in the white matter, with no dual labelling observed with
either CD68+ macrophages (red) or GFAP+ astrocytes. (J) MV-infected (blue) NOGO+ oligodendrocytes (red) are present in the white matter.
(K) No virus spread is observed from MV-infected white matter oligodendrocytes (blue and red) to GFAP+ astrocytes (green). Propidium
iodide (red) was used as nuclear counterstain in (B, D–F); square, neurons; diamond, oligodendrocytes.

• Where are the intracellular sites of MV RNA synthesis Conclusions


in neurons?
• How does the presence of large foreign RNA/protein Collaborative, multidisciplinary research combining
aggregates interfere with the normal functioning of molecular virology, cell and tissue imaging, animal
neurons? models of disease and targeted pathology have radically
Copyright © 2014 Pathological Society of Great Britain and Ireland. J Pathol 2015; 235: 253–265
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
262 M Ludlow et al

changed our understanding of measles pathogenesis. Technology Unit (Queen’s University of Belfast) for
These recent advances, alongside the availability of a their histological expertise. This work was funded
highly efficacious vaccine, make MV a perfect platform by the Medical Research Council (MRC; Grant No.
for comparative studies. Understanding the molecular G0801001) and by ZonMw (Grant No. 91208012) to
basis of attenuation, and dissecting how a vaccine virus WPD and RdS, respectively. The funders had no role in
behaves in vivo compared to the wild-type progenitor, study design, data collection and analysis or decision to
are important. Efforts are being made to move away publish, or in the preparation of the manuscript.
from the standard, empirical approach to live attenuated
vaccine development, in which a clinical isolate is Author contributions
repeatedly passaged in inappropriate cells. Molecular
approaches in which targeted modifications are made WPD and RdS conceived and designed the experiments;
to viruses based on a comprehensive understanding WPD, RdS, ML and SMcQ performed the experiments
of the mechanisms of attenuation are attractive, given and analysed the data; and DM provided pathological
the challenges in licensing new vaccines and the need oversight and commented on existing data; all authors
to ensure vaccine safety. Since measles vaccination were involved in writing the paper; WPD and ML assem-
leads to lifelong immunity, efforts should be made to bled the figures; and all authors approved the submitted
understand the mechanisms of attenuation and assess and published versions.
whether this knowledge can be used for other viruses. At
present we do not know the initial target cells of the MV
vaccine, whether or not it uses non-natural receptors References
in vivo [121,122] or how long the virus persists follow- 1. United Nations. The Millennium Development Report 2009. United
ing the injection of vaccinees. Such basic knowledge Nations: New York, NY, 2009.
is essential and is directly applicable to other live 2. World Health Organization (WHO). Global Measles and Rubella
attenuated vaccines, such as mumps and rubella. Tar- Strategic Plan: 2012–2020. WHO: Geneva, 2012; 1–43.
geted pathology helps to answer these questions and 3. Hirsch A. Handbook of Geographical and Historical Pathology:
the approaches developed to understand the systemic Acute Infective Diseases. New Syndenham Society: London, 1883.
disease are directly applicable to vaccine viruses. Given 4. Keeling MJ, Grenfell BT. Disease extinction and community size:
the genetic stability of MV and its ability to retain extra modeling the persistence of measles. Science 1997; 275: 65–67.
5. Monto AS. Interrupting the transmission of respiratory tract infec-
genes in vivo, it is an ideal platform for the delivery
tions: theory and practice. Clin Infect Dis 1999; 28: 200–204.
of antigens from other pathogens [123,124]. Serious
6. Anderson RM, May RM. Vaccination against rubella and measles:
thought needs to be given to the issues surrounding quantitative investigations of different policies. J Hyg (Lond) 1983;
continued vaccination in a post-MV-eradication world. 90: 259–325.
Issues such as morbillivirus emergence from animals 7. Panum PL. Observations made during the epidempic of measles on
or, less likely but nevertheless possible, the use of the the Faroe Islands in the year 1846. Med Classics 1939; 3: 839–886.
virus as a bioweapon, cannot be ignored. Both should be 8. van Boven M, Kretzschmar M, Wallinga J, et al. Estimation
considered, as it is challenging to maintain vaccination of measles vaccine efficacy and critical vaccination coverage
rates in developed countries when endemic transmission in a highly vaccinated population. J R Soc Interface 2010; 7:
is stopped. In a post-eradication world it is very likely 1537–1544.
that significant numbers of people would opt out of 9. de Swart RL, Duprex WP, Osterhaus AD. Rinderpest eradication:
lessons for measles eradication? Curr Opin Virol 2012; 2: 330–334.
routine MV vaccination. Are these issues threats or
10. OIE. Joint FAO/OIE Committee on Global Rinderpest
opportunities? It could be argued that now is the perfect Eradication. Final report May 2011: http://www.oie.int/fileadmin/
time to invest in understanding both the disease and vac- Home/eng/Media_Center/docs/pdf/Final_Report_May2011.pdf
cine to inform our thinking. Further research is required 11. Drexler JF, Corman VM, Muller MA, et al. Bats host major mam-
in order to understand transmission, the longevity of malian paramyxoviruses. Nat Commun 2012; 3: 796.
immune suppression and the interplay between CD150 12. Baron MD, Goatley L, Barrett T. Cloning and sequence analysis
and PVRL4. This will require strong pathological foun- of the matrix (M) protein gene of rinderpest virus and evidence for
dations and appreciation of the complex lymphotropic, another bovine morbillivirus. Virology 1994; 200: 121–129.
epitheliotropic and neurotropic nature of MV. 13. Keeling MJ. Modelling the persistence of measles. Trends Microbiol
1997; 5: 513–518.
14. Rima BK, Duprex WP. Morbilliviruses and human disease. J Pathol
2006; 208: 199–214.
Acknowledgements 15. Katz SL, Hinman AR. Summary and conclusions: measles elimina-
tion meeting, 16–17 March 2000. J Infect Dis 2004; 189(suppl 1):
S43–47.
We wish to thank Ingrid Allen, Ken Lemon and Bert
16. Centers for Disease Control (CDC). Measles cases and outbreaks:
Rima (Queen’s University of Belfast, Northern Ire-
http://www.cdc.gov/measles/cases-outbreaks.html
land, UK) and Rory de Vries and Albert Osterhaus 17. ECDC. Surveillance report. Measles and Rubella Monitoring. April
(Erasmus MC, Rotterdam, The Netherlands) for helpful 2014: http://www.ecdc.europa.eu/en/publications/Publications/
discussions and support. We acknowledge Geert van measles-rubella-monitoring-april-2014.pdf
Amerongen (Erasmus MC), who provided unparalleled 18. Hall WC, Kovatch RM, Herman PH, et al. Pathology of measles in
zootechnical skills, and the staff of the Tissue Core rhesus monkeys. Vet Pathol 1971; 8: 307–319.

Copyright © 2014 Pathological Society of Great Britain and Ireland. J Pathol 2015; 235: 253–265
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
Measles virus pathology 263

19. MacArthur JA, Mann PG, Oreffo V, et al. Measles in monkeys: an 41. Frenzke M, Sawatsky B, Wong XX, et al. Nectin-4-dependent
epidemiological study. J Hyg (Lond) 1979; 83: 207–212. measles virus spread to the cynomolgus monkey tracheal epithe-
20. van Binnendijk RS, van der Heijden RW, van Amerongen G, et al. lium: role of infected immune cells infiltrating the lamina propria.
Viral replication and development of specific immunity in macaques J Virol 2013; 87: 2526–2534.
after infection with different measles virus strains. J Infect Dis 1994; 42. Beineke A, Puff C, Seehusen F, et al. Pathogenesis and
170: 443–448. immunopathology of systemic and nervous canine distemper.
21. McChesney MB, Miller CJ, Rota PA, et al. Experimental measles. I. Vet Immunol Immunopathol 2009; 127: 1–18.
Pathogenesis in the normal and the immunized host. Virology 1997; 43. Beckford AP, Kaschula RO, Stephen C. Factors associated with
233: 74–84. fatal cases of measles. A retrospective autopsy study. S Afr Med J
22. Zhu YD, Heath J, Collins J, et al. Experimental measles. II. Infec- 1985; 68: 858–863.
tion and immunity in the rhesus macaque. Virology 1997; 233: 44. Akramuzzaman SM, Cutts FT, Wheeler JG, et al. Increased child-
85–92. hood morbidity after measles is short-term in urban Bangladesh. Am
23. Lemon K, de Vries RD, Mesman AW, et al. Early target cells of J Epidemiol 2000; 151: 723–735.
measles virus after aerosol infection of non-human primates. PLoS 45. Rafat C, Klouche K, Ricard JD, et al. Severe measles infection:
Pathog 2011; 7: e1001263. The spectrum of disease in 36 critically ill adult patients. Medicine
24. Takeuchi K, Takeda M, Miyajima N, et al. Stringent requirement for (Baltimore) 2013; 92: e10–e18.
the C protein of wild-type measles virus for growth both in vitro and 46. Shanks GD, Lee SE, Howard A, et al. Extreme mortality after first
in macaques. J Virol 2005; 79: 7838–7844. introduction of measles virus to the polynesian island of Rotuma,
25. El Mubarak HS, Yuksel S, van Amerongen G, et al. Infection of 1911. Am J Epidemiol 2011; 173: 1211–1222.
cynomolgus macaques (Macaca fascicularis) and rhesus macaques 47. Von Pirquet CE. Das Verhalten der kutanen Tuberkulin-reaktion
(Macaca mulatta) with different wild-type measles viruses. J Gen während der Masern. Dtsch Med Wochenschr 1908; 34:
Virol 2007; 88: 2028–2034. 1297–1300.
26. Ludlow M, de Vries RD, Lemon K, et al. Infection of lymphoid 48. Griffin DE, Ward BJ, Jauregui E, et al. Immune activation in
tissues in the macaque upper respiratory tract contributes to the measles. N Engl J Med 1989; 320: 1667–1672.
emergence of transmissible measles virus. J Gen Virol 2013; 94: 49. Mongkolsapaya J, Jaye A, Callan MF, et al. Antigen-specific expan-
1933–1944. sion of cytotoxic T lymphocytes in acute measles virus infection.
27. Ludlow M, Lemon K, de Vries RD, et al. Measles virus infection J Virol 1999; 73: 67–71.
of epithelial cells in the macaque upper respiratory tract is mediated 50. Schneider-Schaulies S, Schneider-Schaulies J. Measles virus-
by subepithelial immune cells. J Virol 2013; 87: 4033–4042. induced immunosuppression. Curr Top Microbiol Immunol 2009;
28. de Vries RD, McQuaid S, van Amerongen G, et al. Measles immune 330: 243–269.
suppression: lessons from the macaque model. PLoS Pathog 2012; 51. Hahm B. Hostile communication of measles virus with host innate
8: e1002885. immunity and dendritic cells. Curr Top Microbiol Immunol 2009;
29. Rima BK, Duprex WP. Molecular mechanisms of measles virus 330: 271–287.
persistence. Virus Res 2005; 111: 132–147. 52. Griffin DE. Measles virus-induced suppression of immune
30. de Vries RD, Mesman AW, Geijtenbeek TB, et al. The pathogenesis responses. Immunol Rev 2010; 236: 176–189.
of measles. Curr Opin Virol 2012; 2: 248–255. 53. de Swart RL, Ludlow M, de Witte L, et al. Predominant infection
31. Warthin AS. Occurrence of numerous large giant cells in the tonsils of CD150+ lymphocytes and dendritic cells during measles virus
and pharyngeal mucosa in the prodromal stage of measles. Arch infection of macaques. PLoS Pathog 2007; 3: e178.
Pathol 1931; 11: 864–874. 54. Noyce RS, Bondre DG, Ha MN, et al. Tumor cell marker PVRL4
32. Bunting CH. The giant-cells of measles. Yale J Biol Med 1950; 22: (Nectin 4) is an epithelial cell receptor for measles virus. PLoS
513–519. Pathog 2011; 7: e1002240.
33. Finkeldey W. Über Riesenzellbefunde in den Gaumenmandeln, 55. Muhlebach MD, Mateo M, Sinn PL, et al. Adherens junction protein
zugleich ein Beitrag zur Histopathologie der Mandelveränderungen nectin-4 is the epithelial receptor for measles virus. Nature 2011;
im Maserninkubationsstadium. Virchows Arch 1931; 281: 323–329. 480: 530–533.
34. Stryker WA. Disseminated giant cell reaction a possible prodrome 56. Reymond N, Fabre S, Lecocq E, et al. Nectin4/PRR4, a new
of measles. Am J Dis Child 59: 468–478. afadin-associated member of the nectin family that trans-interacts
35. Tatsuo H, Ono N, Tanaka K, et al. SLAM (CDw150) is a cellular with nectin1/PRR1 through V domain interaction. J Biol Chem
receptor for measles virus. Nature 2000; 406: 893–897. 2001; 276: 43205–43215.
36. Calpe S, Wang N, Romero X, et al. The SLAM and SAP gene fam- 57. Noyce RS, Delpeut S, Richardson CD. Dog nectin-4 is an epithelial
ilies control innate and adaptive immune responses. Adv Immunol cell receptor for canine distemper virus that facilitates virus entry
2008; 97: 177–250. and syncytia formation. Virology 2013; 436: 210–220.
37. Kiel MJ, Yilmaz OH, Iwashita T, et al. SLAM family receptors 58. Birch J, Juleff N, Heaton MP, et al. Characterization of ovine
distinguish hematopoietic stem and progenitor cells and reveal Nectin-4, a novel peste des petits ruminants virus receptor. J Virol
endothelial niches for stem cells. Cell 2005; 121: 1109–1121. 2013; 87: 4756–4761.
38. Berger SB, Romero X, Ma C, et al. SLAM is a microbial sensor 59. Mandai K, Nakanishi H, Satoh A, et al. Afadin: A novel actin
that regulates bacterial phagosome functions in macrophages. Nat filament-binding protein with one PDZ domain localized at
Immunol 2010; 11: 920–927. cadherin-based cell-to-cell adherens junction. J Cell Biol 1997;
39. de Witte L, Abt M, Schneider-Schaulies S, et al. Measles virus 139: 517–528.
targets DC-SIGN to enhance dendritic cell infection. J Virol 2006; 60. Mandai K, Nakanishi H, Satoh A, et al. Ponsin/SH3P12: an
80: 3477–3486. l-afadin- and vinculin-binding protein localized at cell–cell and
40. Ferreira CS, Frenzke M, Leonard VH, et al. Measles virus infection cell–matrix adherens junctions. J Cell Biol 1999; 144: 1001–1017.
of alveolar macrophages and dendritic cells precedes spread to 61. Brancati F, Fortugno P, Bottillo I, et al. Mutations in PVRL4,
lymphatic organs in transgenic mice expressing human signaling encoding cell adhesion molecule nectin-4, cause ectodermal
lymphocytic activation molecule (SLAM, CD150). J Virol 2010; 84: dysplasia–syndactyly syndrome. Am J Hum Genet 2010; 87:
3033–3042. 265–273.

Copyright © 2014 Pathological Society of Great Britain and Ireland. J Pathol 2015; 235: 253–265
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
264 M Ludlow et al

62. Takano A, Ishikawa N, Nishino R, et al. Identification of nectin-4 83. Hardie DR, Albertyn C, Heckmann JM, et al. Molecular characteri-
oncoprotein as a diagnostic and therapeutic target for lung cancer. sation of virus in the brains of patients with measles inclusion body
Cancer Res 2009; 69: 6694–6703. encephalitis (MIBE). Virol J 2013; 10: 283.
63. Nabih ES, Abdel Motaleb FI, Salama FA. The diagnostic efficacy 84. Freeman AF, Jacobsohn DA, Shulman ST, et al. A new complica-
of nectin 4 expression in ovarian cancer patients. Biomarkers 2014; tion of stem cell transplantation: measles inclusion body encephali-
19: 1–7. tis. Pediatrics 2004; 114: e657–660.
64. Derycke MS, Pambuccian SE, Gilks CB, et al. Nectin 4 overexpres- 85. Bitnun A, Shannon P, Durward A, et al. Measles inclusion-body
sion in ovarian cancer tissues and serum: potential role as a serum encephalitis caused by the vaccine strain of measles virus. Clin
biomarker. Am J Clin Pathol 2010; 134: 835–845. Infect Dis 1999; 29: 855–861.
65. Fabre-Lafay S, Garrido-Urbani S, Reymond N, et al. Nectin-4, 86. Spalke G, Eschenbach C. Infantile cortical measles inclusion body
a new serological breast cancer marker, is a substrate for tumor encephalitis during combined treatment of acute lymphoblastic
necrosis factor-α-converting enzyme (TACE)/ADAM-17. J Biol leukemia. J Neurol 1979; 220: 269–277.
Chem 2005; 280: 19543–19550. 87. Greenfield JG. Encephalitis and encephalomyelitis in England and
66. Pavlova NN, Pallasch C, Elia AE, et al. A role for PVRL4-driven Wales during the last decade. Brain 1950; 73: 141–166.
cell–cell interactions in tumorigenesis. Elife 2013; 2: e00358. 88. Connolly JH, Allen IV, Hurwitz LJ, et al. Measles-virus antibody
67. Mateo M, Navaratnarajah CK, Syed S, et al. The measles virus and antigen in subacute sclerosing panencephalitis. Lancet 1967; 1:
hemagglutinin β-propeller head β4–β5 hydrophobic groove governs 542–544.
functional interactions with nectin-4 and CD46 but not those with 89. Gagnon A, Bouchard RW. Fulminating adult-onset subacute scle-
the signaling lymphocytic activation molecule. J Virol 2013; 87: rosing panencephalitis in a 49 year-old man. Arch Neurol 2003; 60:
9208–9216. 1160–1161.
68. Delpeut S, Noyce RS, Richardson CD. The V domain of dog 90. Freeman JM. The clinical spectrum and early diagnosis of Dawson’s
PVRL4 (nectin-4) mediates canine distemper virus entry and virus encephalitis, with preliminary notes on treatment. J Pediatr 1969;
cell-to-cell spread. Virology 2014; 454–455: 109–117. 75: 590–603.
69. Katayama Y, Hotta H, Nishimura A, et al. Detection of measles 91. Connolly JH, Allen IV, Hurwitz LJ, et al. Subacute sclerosing
virus nucleoprotein mRNA in autopsied brain tissues. J Gen Virol panencephalitis. Clinical, pathological, epidemiological, and viro-
1995; 76: 3201–3204. logical findings in three patients. Qu J Med 1968; 37: 625–644.
70. Katayama Y, Kohso K, Nishimura A, et al. Detection of measles 92. Miller C, Farrington CP, Harbert K. The epidemiology of subacute
virus mRNA from autopsied human tissues. J Clin Microbiol 1998; sclerosing panencephalitis in England and Wales 1970–1989. Int J
36: 299–301. Epidemiol 1992; 21: 998–1006.
71. Gibbs EL, Grossman HJ, Spies HW. Electroencephalographic stud- 93. Tan E, Namer IJ, Ciger A, et al. The prognosis of subacute scleros-
ies on children having measles with no clinical evidence of involve- ing panencephalitis in adults. Report of 8 cases and review of the
ment of the central nervous system. Pediatrics 1956; 18: 556–560. literature. Clin Neurol Neurosurg 1991; 93: 205–209.
72. Fisher DL, Defres S, Solomon T. Measles-induced encephalitis. Qu 94. Mahadevan A, Vaidya SR, Wairagkar NS, et al. Case of
J Med 2014; DOI: 10.1093/qjmed/hcu113. fulminant-SSPE associated with measles genotype D7 from
73. Buchanan R, Bonthius DJ. Measles virus and associated central India: an autopsy study. Neuropathology 2008: 28: 621–626.
nervous system sequelae. Semin Pediatr Neurol 2012; 19: 107–114. 95. Bellini WJ, Rota JS, Lowe LE, et al. Subacute sclerosing panen-
74. Javed A, Khan O. Acute disseminated encephalomyelitis. Handb cephalitis: more cases of this fatal disease are prevented by measles
Clin Neurol 2014; 123: 705–717. immunization than was previously recognized. J Infect Dis 2005;
75. Elhassanien AF, Aziz HA. Acute demyelinating encephalomyelitis: 192: 1686–1693.
clinical characteristics and outcome. J Pediatr Neurosci 2013; 8: 96. Manning L, Laman M, Edoni H, et al. Subacute sclerosing panen-
26–30. cephalitis in papua new guinean children: the cost of continuing
76. Mohsen H, Abu Zeinah GF, Elsotouhy AH, et al. Acute dissem- inadequate measles vaccine coverage. PLoS Negl Trop Dis 2011;
inated encephalomyelitis following infectious mononucleosis in a 5: e932.
toddler. BMJ Case Rep 2013; DOI: 10.1136/bcr-2013-010048. 97. Takasu T, Mgone JM, Mgone CS, et al. A continuing high inci-
77. Miller HG, Stanton JB, Gibbons JL. Acute disseminated dence of subacute sclerosing panencephalitis (SSPE) in the East-
encephalomyelitis and related syndromes. Br Med J 1957; 1: ern Highlands of Papua New Guinea. Epidemiol Infect 2003; 131:
668–672. 887–898.
78. Johnson RT, Griffin DE, Hirsch RL, et al. Measles encephalomyeli- 98. Schonberger K, Ludwig MS, Wildner M, et al. Epidemiology of
tis – clinical and immunologic studies. N Engl J Med 1984; 310: subacute sclerosing panencephalitis (SSPE) in Germany from 2003
137–141. to 2009: a risk estimation. PLoS One 2013; 8: e68909.
79. Nardone R, Golaszewski S, Trinka E, et al. Acute disseminated 99. Dawson JR. Cellular inclusions in cerebral lesions of lethargic
encephalomyelitis preceding measles exanthema. J Child Neurol encephalitis. Am J Pathol 1933; 9: 7–16.
2011; 26: 1590–1592. 100. Dawson JR. Cellular inclusions in cerebral lesions of epidemic
80. Mustafa MM, Weitman SD, Winick NJ, et al. Subacute measles encephalitis, second report. Arch Neur Psych 1934; 31: 685–700.
encephalitis in the young immunocompromised host: report of two 101. Bouteille M, Fontaine C, Vedrenne CL, et al. Sur un cas
cases diagnosed by polymerase chain reaction and treated with d’encephalite subaigue a inclusions. Étude anatomclinique et
ribavirin and review of the literature. Clin Infect Dis 1993; 16: ultrastructurelle. Rev Neurol (Paris) 1965; 118: 454–458.
654–660. 102. Cattaneo R, Schmid A, Rebmann G, et al. Accumulated measles
81. Agamanolis DP, Tan JS, Parker DL. Immunosuppressive measles virus mutations in a case of subacute sclerosing panencephalitis:
encephalitis in a patient with a renal transplant. Arch Neurol 1979; interrupted matrix protein reading frame and transcription alter-
36: 686–690. ation. Virology 1986; 154: 97–107.
82. Aicardi J, Goutieres F, Arsenio-Nunes ML, et al. Acute measles 103. Cattaneo R, Schmid A, Eschle D, et al. Biased hypermutation and
encephalitis in children with immunosuppression. Pediatrics 1977; other genetic changes in defective measles viruses in human brain
59: 232–239. infections. Cell 1988; 55: 255–265.

Copyright © 2014 Pathological Society of Great Britain and Ireland. J Pathol 2015; 235: 253–265
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com
Measles virus pathology 265

104. Cattaneo R, Schmid A, Spielhofer P, et al. Mutated and hypermu- 114. Isaacson SH, Asher DM, Godec MS, et al. Widespread, restricted
tated genes of persistent measles viruses which caused lethal human low-level measles virus infection of brain in a case of subacute
brain diseases. Virology 1989; 173: 415–425. sclerosing panencephalitis. Acta Neuropathol 1996; 91: 135–139.
105. Schmid A, Spielhofer P, Cattaneo R, et al. Subacute sclerosing 115. Iwasaki Y, Koprowski H. Cell-to-cell transmission of virus in the
panencephalitis is typically characterized by alterations in the fusion central nervous system. I. Subacute sclerosing panencephalitis. Lab
protein cytoplasmic domain of the persisting measles virus. Virology Invest 1974; 31: 187–196.
1992; 188: 910–915. 116. Seki F, Yamada K, Nakatsu Y, et al. The SI strain of measles virus
106. Herndon RM, Rubinstein LJ. Light and electron microscopy obser- ferived from a patient with subacute scleorosing panencephalitis
vations on the development of viral particles in the inclusions of possesses typical genome alterations and unique amino acid changes
Dawson’s encephalitis (subacute sclerosing panencephalitis). Neu- that modulate receptor specificity and reduce membrane fusion
rology 1968; 18: 8–20. activity. J Virol 2011; 85: 11871–11882.
107. Brown HR, Goller NL, Rudelli RD, et al. Postmortem detection of 117. Mesquita R, Castanos-Velez E, Biberfeld P, et al. Measles virus
measles virus in non-neural tissues in subacute sclerosing panen- antigen in macrophage/microglial cells and astrocytes of subacute
cephalitis. Ann Neurol 1989; 26: 263–268. sclerosing panencephalitis. Acta Pathol Microbiol Immunol Scand
108. Fournier JG, Tardieu M, Lebon P, et al. Detection of measles virus 1998; 106: 553–561.
RNA in lymphocytes from peripheral-blood and brain perivascu- 118. Kumanishi T, In S. SSPE: immunohistochemical demonstration
lar infiltrates of patients with subacute sclerosing panencephalitis. of measles virus antigen(s) in paraffin sections. Acta Neuropathol
N Engl J Med 1985; 313: 910–915. 1979; 48: 161–163.
109. Fournier JG, Gerfaux J, Joret AM, et al. Subacute sclerosing panen- 119. Paula-Barbosa MM, Cruz C. Nerve cell fusion in a case of subacute
cephalitis: detection of measles virus sequences in RNA extracted sclerosing panencephalitis. Ann Neurol 1981; 9: 400–403.
from circulating lymphocytes. Br Med J (Clin Res Ed) 1988; 296: 120. Esiri MM, Oppenheimer DR, Brownell B, et al. Distribution of
684. measles antigen and immunoglobulin-containing cells in the CNS
110. Kirk J, Zhou AL, McQuaid S, et al. Cerebral endothelial cell infec- in subacute sclerosing panencephalitis (SSPE) and atypical measles
tion by measles virus in subacute sclerosing panencephalitis: ultra- encephalitis. J Neurol Sci 1982; 53: 29–43.
structural and in situ hybridization evidence. Neuropathol Appl Neu- 121. Naniche D, Varior-Krishnan G, Cervoni F, et al. Human membrane
robiol 1991; 17: 289–297. cofactor protein (CD46) acts as a cellular receptor for measles virus.
111. McQuaid S, Kirk J, Zhou AL, et al. Measles virus infection of cells J Virol 1993; 67: 6025–6032.
in perivascular infiltrates in the brain in subacute sclerosing panen- 122. Dörig RE, Marcil A, Chopra A, et al. The human CD46 molecule
cephalitis: confirmation by non-radioactive in situ hybridization, is a receptor for measles virus (Edmonston strain). Cell 1993; 75:
immunocytochemistry and electron microscopy. Acta Neuropathol 295–305.
1993; 85: 154–158. 123. Escriou N, Callendret B, Lorin V, et al. Protection from SARS
112. McQuaid S, Cosby SL. An immunohistochemical study of the coronavirus conferred by live measles vaccine expressing the spike
distribution of the measles virus receptors, CD46 and SLAM, in glycoprotein. Virology 2014; 452–453: 32–41.
normal human tissues and subacute sclerosing panencephalitis. Lab 124. Brandler S, Ruffie C, Najburg V, et al. Pediatric measles vaccine
Invest 2002; 82: 403–409. expressing a dengue tetravalent antigen elicits neutralizing antibod-
113. Allen IV, McQuaid S, McMahon J, et al. The significance of ies against all four dengue viruses. Vaccine 2010; 28: 6730–6739.
measles virus antigen and genome distribution in the CNS in SSPE 125. De Vries R, de Swart RL. Measles immune suppression: functional
for mechanisms of viral spread and demyelination. J Neuropathol impairment or numbers game. PLoS Pathog, doi: 10.1371/journal.
Exp Neurol 1996; 55: 471–480. ppat.1004482 (in press).

Copyright © 2014 Pathological Society of Great Britain and Ireland. J Pathol 2015; 235: 253–265
Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com

Potrebbero piacerti anche