You are on page 1of 13

REVIEWS

Organ-specific protection mediated


by cooperation between vascular
and epithelial barriers
Ilaria Spadoni1, Giulia Fornasa1 and Maria Rescigno1,2
Abstract | Immune privilege is a complex process that protects organs from immune-mediated
attack and damage. It is accomplished by a series of cellular barriers that both control immune
cell entry and promote the development of tolerogenic immune cells. In this Review, we describe
the vascular endothelial and epithelial barriers in organs that are commonly considered to be
immune privileged, such as the brain and the eye. We compare these classical barriers with
barriers in the intestine, which share features with barriers of immune-privileged organs, such as
the capacity to induce tolerance and to protect from external insults. We suggest that when
intestinal barriers break down, disruption of other barriers at distant sites can ensue, and this may
underlie the development of various neurological, metabolic and intestinal disorders.

Inflammatory bowel diseases The original concept of ‘immune privilege’ was coined to to antigens and leukocytes10. In many organs, such as
(IBDs). Chronic disorders of the explain the failure of certain tissues to reject implanted the intestine and brain, these barriers work together
intestine characterized by grafts1. Many tissues and organs are considered to be to form a complex protective layer. This is necessary
severe inflammation and immune privileged — they include the brain, retina, because the intestine has to preserve the microbiota
mucosal tissue destruction.
There are two main forms:
testis and feto–maternal interface. However, this concept but, at the same time, prevent it from entering the
Crohn’s disease, which is a of immune privilege has recently been extended to include systemic circulation11. In the central nervous system
granulomatous segmental sites of immune tolerance induction, such as the intestine, (CNS), endothelial and epithelial barriers allow the
inflammation affecting any part which is tolerant to food antigens and the trillions of com- immunosurveillance of the CNS without the risk of
of the intestine, and ulcerative
mensal microorganisms2–6, and tumours7. This extended perturbing the homeostasis of the brain parenchyma12.
colitis, which is a mucosal
inflammation involving the
definition of immune privilege to include the intestine These barriers in the intestine and brain have similar-
rectum and extending for a does not refer to an exclusion of immune cells but rather ities and differences in their anatomy, properties and
variable distance along the to the capacity to avoid the induction of systemic immu- development, and we review recent efforts that have
colon. nity against dietary antigens and the microbiota. In this revealed a functional connection between vascular
respect, immune privilege seems to be a much more barriers at different sites of the body, especially the
complex process than previously appreciated and can intestine and brain. The use of experimental mouse
involve different mechanisms depending on the organ. models, together with genomics, metagenomics and
It is established by at least two mechanisms: a passive metabolomics, has shed light on the mechanisms that
physical restriction of leukocyte access across vas­cular underlie barrier development and maintenance and
barriers, such as the blood–brain barrier (BBB)8,9, and has highlighted the role of the intestinal microbiota
an active mechanism that involves immune-­regulatory in shaping the properties of the intestinal and brain
1
Department of Experimental
processes, such as those at the blood–cerebrospinal barriers. Indeed, intestinal microorganisms can influ-
Oncology, European fluid (CSF) interface and the intestinal epithelial barrier ence angiogenesis13,14, modulate intestinal function15,
Institute of Oncology, (IEB), that ensure the skewing of immune cells towards a shape BBB properties and influence behaviour 16–19. The
20139 Milan, Italy. regulatory phenotype. microbiota is also involved in the establishment of an
2
Dipartimento di Scienze
In this Review, we broaden the model of immune efficient vascular barrier in other organs, such as the
della Salute, Università degli
Studi di Milano, gates proposed by Shechter et al.10 and highlight the testis20 and eye21. Thus, it is evident that tissue barriers
20122 Milan, Italy. role of the intestine in establishing immune privilege, are dynamic during the steady state; however, much of
Correspondence to M.R. both in the intestine and in other tissues. We analyse the knowledge about barrier modifications comes from
maria.rescigno@ieo.it the function of barrier systems, which consist of vas- disease conditions, such as neurological disorders,
doi:10.1038/nri.2017.100 cular barriers that restrict the trafficking of immune meta­b olic disorders and inflammatory bowel diseases
Published online 4 Sep 2017 cells and of epithelial barriers that are more permissive (IBDs). We first describe the nature of the different

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 1


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

barriers and then examine how several pathologies are and will undergo apoptosis. By contrast, upon antigen
linked to the disruption of these barriers, with a focus recognition, activated T cells become encephalitogenic
on how barriers in different organs can influence each and start to produce pro-inflammatory cytokines that
other in pathological settings. alter BBB properties and trigger the upregulation of the
expression of chemokines that recruit myeloid cells.
The brain barriers Myeloid cells are responsible for the cleavage of
Anatomy of the BBB. The CNS is protected by differ- dys­troglycan at the membrane of astrocyte endfeet, and
ent layers. The outermost barrier, beneath the inner this process allows the subsequent infiltration of immune
surface of the skull, is formed by the meninges, namely, cells across the glia limitans into the CNS parenchyma and
the dura mater, arachnoid mater and pia mater, which promotes the development of experimental autoimmune
surround the brain and the CSF. This layer is permissive enceph­alomyelitis (EAE)12, a mouse model of multiple
to immune cell trafficking (FIG. 1a). Underneath the pia sclerosis (MS).
mater, the glia limitans forms a barrier that restricts the
trafficking of T cells. This barrier consists of astrocyte Formation of the BBB. Early studies of quail–chick
endfeet and the parenchymal basement membrane that transplantation demonstrated that the barrier
covers blood vessels and the brain parenchymal surface characteristics of endothelial cells are not intrinsic and
(FIG. 1a). Deeper within the brain, two further barri- are induced by the CNS environment 32,33. Indeed, newly
ers are present: the BBB, which involves parenchymal formed sprouting vessels express junctional proteins and
capill­aries, and the blood–CSF barrier (BCSFB), which nutrient transporters, show high levels of transcytosis
is localized at the choroid plexus in the brain ventricles. and express leukocyte adhesion molecules 25. The
These two barriers have different cellular compositions characteristics of the BBB are induced and maintained by
and permeability characteristics; the BCSFB establishes the interaction of brain endothelial cells with pericytes,
Meninges
Vascularized tissue membranes
a much tighter barrier to soluble tracers and higher elec- astrocytes and components of the extracellular matrix,
that envelop superficial central trical resistance than the BBB. They are also character- which together are referred to as the neurovascular unit
nervous system areas and ized by differences in permissiveness to immune cells, (NVU) (FIG. 1b).
enclose the parenchyma. conferring diverse degrees of immune privilege to these Formation of the BBB starts when endothelial pro-
The meninges are composed of
regions of the brain22; tissue grafts transplanted into genitor cells extend from the perineural vascular plexus
three layers: the dura mater
(outermost, beneath the skull), cerebral ventricles are rejected, but they are not rejected into the embryonic neuroectoderm through a process
the arachnoid mater and the if transplanted into parenchymal tissue23,24. that is guided by neural progenitor cells that secrete par-
pia mater (the innermost layer, The BBB is present across the capillaries and acrine factors34 such as vascular endothelial growth fac-
which is proximal to the post-capillary venules of the CNS25, the core element of tor (VEGF)35 and WNT ligands. Embryonic deletion of
parenchyma).
which is the blood microvasculature that is composed the genes encoding components of the WNT–β‑catenin
Astrocyte of specialized endothelial cells. The endothelial cells of signalling pathway is lethal, as endothelial cells fail to
A type of glial cell that is found the BBB are characterized by low rates of pinocytosis, a extend into the CNS parenchyma and instead form
in the vertebrate brain and is lack of fenestrations and continuous intercellular junc- large haemorrhagic aggregates that lack expression of
named for its characteristic
tional complexes formed by tight junction and adherens BBB-specific genes34,36. However, postnatal in­­activation
star-like shape. Astrocytes
provide both mechanical and junction proteins that limit the paracellular leakage of of the β‑catenin pathway in endothelial cells leads to
metabolic support for neurons, molecules. The passage of molecules that are necessary the reduced expression of claudin 3, a component of
thereby regulating the for the CNS and for the clearance of toxic agents is medi- tight junctions, and increased expression of plasma-
environment in which neurons ated by several specific transporters25. However, the infil- lemma vesicle-associated protein (PLVAP), a marker
function.
tration of immune cells from the blood into the healthy of vesi­cular transport, and these changes correlate with
Experimental autoimmune brain is limited by the endothelial cells of the BBB owing vascular leakage and reduced BBB maturation37. Thus,
encephalomyelitis to their low expression of leukocyte adhesion molecules the canonical WNT–β‑catenin signalling pathway is
(EAE). An experimental model and the production of mediators, such as sonic hedgehog fundamental for brain angiogenesis and for BBB matur­
for the human disease multiple
(SHH), that limit immune cell adhesion, migration and ation. In addition to the β‑catenin pathway, adhesion G
sclerosis. Autoimmune disease
is induced in experimental pro-inflammatory cytokine production26. This special- protein-coupled receptor A2 (ADGRA2; also known as
animals by immunization with ized function of the parenchymal endothelium resides in GPR124) is expressed by developing brain endothelial
myelin or peptides derived the post-capillary venules, the vascular segment in which cells and is necessary for normal angiogenesis and for
from myelin. The animals the endothelial and parenchymal basement membranes barrier development 38–40.
develop a paralytic disease
with inflammation and
create a perivascular space that can accommodate rare After capillary formation, the second stage
demyelination in the brain and antigen-presenting cells (APCs)27–30. Interestingly, CNS- of BBB development is mediated by the interaction of
spinal cord. resident APCs have been shown to be able to sample CNS endo­thelial cells with pericytes and astrocytes, which
antigens and activate CD4+ and CD8+ T cells. The infiltra- promote the ‘sealing’ of the BBB. Mice deficient
Pericytes
tion of activated T cells into the CNS is restricted during in platelet-­derived growth factor receptor‑β (Pdgfrb)
Cells that are embedded in the
vascular basement membrane the steady state31; however, they can cross the BBB in the or platelet-derived growth factor subunit B (Pdgfb)
of microvessels. They make absence of neuroinflammation and independent of their completely lack CNS pericytes, exhibit endothelial cell
close contact with endothelial antigen specificity. Activated T cells can enter the CNS hyperplasia and show increased vessel diameter and vas-
cells, and this interaction is through subpial venules or subarachnoid veins, or they cular permeability, which results in micro­haemorrhages
essential for the maintenance
of vessel function, as well as for
can reach the lepto­meningeal spaces through the choroid and perinatal lethality 41,42. Moreover, the absence of peri-
the regulation of angiogenesis plexus. If T cells do not recognize their cognate antigen cytes during brain development has been associated with
and vascular remodelling. presented by APCs, they do not cross the glia limitans increased expression of markers of a more permeable

2 | ADVANCE ONLINE PUBLICATION www.nature.com/nri


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

a Meninges and BLMB

Skull bone

Dura mater

Arachnoid mater
Subarachnoid space b BBB: exclusive barrier
BLMB
Neuron
Pia mater
Astrocyte
Glia limitans APOE
Microglia

Tight junction Brain SHH


parenchyma

ANG1

RA

Pericyte

Basement
membrane

Meninges

Tight
junction
Endothelium

Post-capillary Parenchymal c BCSFB: regulatory barrier


Ventricle capillary
Brain parenchyma

CSF Cuboidal Tight Ependymal cells


epithelium junction

Basal membrane

Fenestrated capillary
Choroid plexus stroma

Figure 1 | Cellular components of brain barriers. a | The meninges sealed by complex junctions mainly formed NaturebyReviews
claudin| 5, astrocyte
Immunology
(comprising the dura mater, arachnoid mater and pia mater) form the endfeet that encircle the abluminal side of the vasculature and pericytes.
outermost barrier that protects the brain parenchyma. The meningeal Together, these cellular components of the BBB form the functional unit
microvessels in the subarachnoid space, which is located between the called the neurovascular unit (NVU). Neurons and microglia are
arachnoid mater and pia mater, are composed of a non-fenestrated considered to be part of the NVU, as they can influence barrier function.
endothelium that contains tight junctions but is not ensheathed by Sonic hedgehog (SHH), angiopoietin 1 (ANG1), apolipoprotein E (APOE)
astroglial endfeet. The pia mater, which covers the surface of the central and retinoic acid (RA) are produced by astrocytes and promote BBB
nervous system (CNS), surrounds these blood vessels to form the blood– integrity. c | The cuboidal epithelial cells of the choroid plexus form the
leptomeningeal barrier (BLMB), which is permeable to solutes and blood–cerebrospinal fluid (CSF) barrier (BCSFB). These epithelial cells are
immune cells. b | The blood–brain barrier (BBB) is necessary for brain connected by tight junctions and have a polarized distribution of
homeostasis and neuron preservation, as it controls the transport of numerous transporters. The endothelium of the choroid plexus does not
solutes and fluids and the extravasation of immune cells into the CNS form a barrier but is permeable to allow the delivery of water from the
parenchyma. The BBB consists of capillary endothelial cells that are blood to the epithelium, which is necessary for CSF production.

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 3


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

vascular barrier, such as PLVAP and leukocyte adhe- homeostatic conditions, the BRB prevents the entry
sion molecules. By contrast, features of a ‘tight’ barrier, of pathogens, allergens and any kind of protein into
such as expression of junctional proteins and transport- the eye. The second element of protection is provided
ers, are not influenced by a lack of pericytes42,43. Pericyte by a strong anti-inflammatory environment and a
coverage is also important for the regulation of BBB skewed immune response to antigens that prevents
integrity in adults; CNS pericyte depletion during age- inflammatory reactions.
ing leads to a reduction in brain microcirculation and
an increase in neuro­inflammatory responses, which Anatomy of the BRB. Similar to the barriers of the
precipitate changes in neuronal structure and behav- CNS, the BRB comprises two physical barriers with
iour 42–44. An additional role of pericytes in the NVU distinct locations, cell compositions and immune-­
is the establishment of astrocyte–­vessel contact and skewing properties. The inner BRB, which is located
astrocyte polarization during the postnatal period42,43. in the inner layers of the neural retina near the vit­
Pericytes induce the polarization of astrocyte endfeet reous body, is a true endothelial barrier. It is formed by
that enclose neuronal processes and the ablu­minal side a specialized non-fenestrated endothelium surrounded
of blood vessels. Moreover, many of the factors secreted by astrocytes, Müller cells and pericytes, which together
by astrocytes have been identified as promoters of BBB form the retinal NVU (reviewed in REF. 59) (FIG. 2a).
integrity during postnatal development and in adult- Astrocytes60,61, Müller cells62,63 and pericytes60,64,65 are
hood. One of these is SHH, which induces the expression thought to contribute to the proper functioning of
of junctional proteins, reduces vascular permeability and the inner BRB. Indeed, in non-primate mammals, the
promotes immune quiescence by downregulating the development of the retinal vasculature starts after birth
expression of pro-inflammatory cytokines and reduc- when it is immediately ensheathed by pericytes64 and
ing leukocyte adhesion and extravasation26. In addition, astrocyte endfeet 61. However, it is still unclear how the
astrocytes enhance barrier properties through the secre- retinal endothelium acquires barrier properties. It has
tion of angiopoietin 1 (REF. 45), apolipoprotein E46–48 and been shown that although the major cellular compo-
retinoic acid49 (FIG. 1b). nents of the BRB are in close contact immediately after
birth, the endothelium acquires barrier characteristics
Components of the BCSFB. Unlike the BBB, which is a only after 10 days64,65; whether this is a result of micro-
true vascular barrier, the BCSFB in the choroid plexus biota colonization of the intestine remains to be estab-
is an epithelium-based barrier 10 that reflects its ability lished. Studies of transgenic animal models in which
to secrete CSF (reviewed in REFS 50–52). The ventri­ pericytes, astrocytes or Müller cells have been depleted
cular choroid plexus is lined by a polarized monolayer of indicate that all these cell types are key regulators of
cuboidal epithelial cells that are joined by junctional com- the induction of BRB properties. For example, mice
plexes, which are mainly composed of claudins 1, 2, 3, 9, 11 that lack pericyte coverage of retinal endothelial cells
and 19 (REFS 53–55). The ventricular epithelium surrounds show decreased expression of claudin 5 and increased
a network of fenestrated capillaries that are not ensheathed expression of PLVAP compared with control mice,
by astrocyte endfeet 56 (FIG. 1c), unlike the endothelium which suggests increased paracellular and trans­cellular
in the parenchyma. This permeable endothelium is transport 64,65. Further evidence for the importance of
necessary for the delivery of water from the blood to the pericytes in the maintenance of a functional BRB is
epithelium for CSF production56 and also for immuno­ provided by studies on diabetic retinopathy, in which
Fenestrated capillaries surveillance of the CNS10. Immunosurveillance is achieved a reduction in pericyte numbers caused increased
A non-continuous vascular bed
characterized by the presence
by an afferent and efferent connection with cervical and permeability of retinal vessels to injected tracers and
of pore-like subcellular lumbar lymph nodes, to which parenchymal antigens are ensuing inflammation65. Indeed, pericyte depletion
structures, or fenestrae, that drained (reviewed in REF. 12). induced the expression of pro-inflammatory genes,
are responsible for transcellular Thus, the CNS is protected by two barriers, namely, such as Tnf, Icam1, Il6 and Ccl2, by endothelial cells,
exchange of molecules.
the endothelial BBB and the epithelial BCSFB, that inducing perivascular infiltration of macrophages
Fenestrated endothelia are
located in the intestine, together provide both passive and active protection. responsible for retinal vessel damage. By contrast,
pancreas, endocrine glands, Similar strategies of protection can also be found in other macrophage depletion in the mouse model of diabetic
glomeruli of the kidney and organs, such as the eye and intestine, as described below. retinopathy suppressed retinal oedema and promoted
liver sinusoids. the recovery of vessel organization65.
Müller cells
The blood–retinal barriers In addition to the inner endothelial-based barrier,
The major type of glial cell that Immune privilege of the anterior chamber of the eye the BRB has an outer epithelial layer that is permissive
is found in the retina. These is achieved by distinct, overlapping mechanisms of to transcellular transport. It is composed of the retinal
cells contribute to retinal protection, both passive and active57. The first ele- pigment epithelium, which separates the outer neural
homeostasis and function and
ments of protection are a physical separation between retina from the fenestrated choriocapillaries (FIG. 2b). The
regulate the tightness of the
blood–retinal barrier. antigens in the eye and the immune system owing retinal pigment epithelium has an important role in reg-
to the blood–­retinal barrier (BRB) and a paucity of ulating the homeostasis of the retina, as it is involved
Diabetic retinopathy efferent lymphatic vessels; these elements ensure in the transport of glucose, lactose, retinol (which are
A condition affecting people that the anterior chamber drains into the blood and necessary for visual function), fatty acids and major
with diabetes that causes
progressive damage to the
not the lymph. The anterior chamber is connected com­ponents of photoreceptors and in the removal of
retina, the light-sensitive tissue directly to the blood circulation via Schlemm’s canal, metabolic waste and water from the subretinal space
at the back of the eye. which is a lymphatic-like vessel58. Accordingly, under (reviewed in REFS 66,67).

4 | ADVANCE ONLINE PUBLICATION www.nature.com/nri


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Aqueous
humour

b Outer BRB: regulatory barrier


a Inner BRB: true barrier

Müller cell Rods and cones Sub-retinal


space

Tight
junction
Pericyte Retinal
Basement pigment
membrane epithelial
cell
CD95L PDL1 Galectin 1 TGFβ IL-10
Bruch’s
membrane
Fenestrated
capillary
Tight
Endothelial junction Choroid plexus
cell
Sclera

Figure 2 | The blood–retinal barrier. The neural retina is equipped with barrier systems that regulate ion, water and
Nature Reviews
molecule flux out of the retina. The blood–retinal barrier (BRB) can be separated into two components: | Immunology
the inner BRB
formed by tight junctions that seal neighbouring capillary endothelial cells (part a) and the outer BRB, which has tight
junctions that restrict paracellular trafficking between retinal pigment epithelial cells (part b). Both layers are fundamental
for maintaining the immune-privileged status of the eye and for regulating retinal homeostasis and visual function.
PDL1, programmed cell death 1 ligand 1; TGFβ, transforming growth factor‑β.

Immune regulation by the BRB. In addition to being create an environment that dampens immune and
a physical barrier to ensure homeostasis of the eye, inflammatory responses within the eye. The inhibi-
the BRB creates an immunosuppressive milieu that tory factors include macrophage migration inhibitory
inhibits the activation of immune cells as they cross factor (MIF), IL‑10, retinoic acid, immuno­suppressive
the barrier. Hence, similar to the BBB and BCSFB, neuropeptides, such as α‑melanocyte-stimulating
Aqueous humour
The clear immunosuppressive
the inner and outer BRB function together to pro- hormone and vasoactive intestinal peptide, and
and anti-inflammatory fluid mote immune privilege. The outer BRB also expresses complement regulatory proteins70–72.
that fills the anterior chamber immunoregulatory molecules that inhibit lympho- The immune-privileged status of the eye is also
of the eye. cyte activation, such as transforming growth factor‑β achieved by a systemic component known as anterior
(TGFβ), cytotoxic T lymphocyte antigen 4 (CTLA4), chamber-associated immune deviation (ACAID), which
Anterior
chamber-associated thrombospondin 1, programmed cell death 1 ligand mediates the downregulation of cellular and humoral
immune deviation 1 (PDL1) and galectin 1, and molecules that promote immune responses to antigens that are encountered in
(ACAID). A form of eye-derived apoptosis, such as CD95 ligand (also known as FAS the anterior chamber of the eye73. Specifically, an anti-
tolerance in which T helper 1 ligand) and TNF-related apoptosis-inducing ligand gen experimentally injected into the anterior chamber
(TH1)- and TH2‑mediated
immunity is suppressed but
(TRAIL; also known as TNFSF10)10,68,69. The retinal or released after injury induces a skewed response that
non-inflammatory adaptive pigment epithelium of the BRB secretes immuno- includes the generation of antigen-specific CD4+ and
immune effectors are present. modulatory mediators into the aqueous humour to CD8+ regulatory T cells and non-complement-binding

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 5


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

antibodies at the expense of potentially destruc- in preserving intestinal epithelial and endothelial bar-
tive T helper 1 (TH1) cell, TH2 cell and delayed-type rier integrity. Transgenic mice that lack enteric glial cells
hypersensitivity inflammatory reactions74. die from intestinal disease owing to increased epithelial
permeability and microvascular disturbances that result
The intestinal barriers in the spread of bacteria into the blood circulation80,81.
Structural barriers. The gastrointestinal tract is a com- Intestinal glial cells could support barrier function by
plex ecosystem that contains the microbiota, which has releasing soluble factors, such as S-nitrosoglutathione,
evolved together with the host species and established a which is known to restore epithelial barrier integrity in
mutualistic relationship. The microbiota is required by mice that lack enteric glial cells82. Interestingly, it has
the host for harvesting energy from food by de­­grading been shown that the formation and maintenance of the
complex molecules and for forming a first barrier of network of enteric glial cells in the lamina propria are
protection against potential pathogens, whereas the guided by the microbiota83. The microbiota is also in­­
host provides nutrients and a niche to the microbiota75. dispensable for angiogenesis in the intestine13; therefore,
Despite this mutualistic relationship, the presence of it is possible that it could influence the GVB directly and
the microbiota poses immense health challenges to indirectly through enteric glial cells, although this needs
the host. Accordingly, several strategies have evolved to be formally demonstrated.
in the intestine to protect the host from the threat of Maintenance of GVB integrity, as for BBB integ-
infection and, at the same time, allow the host to toler- rity 34,36, is regulated by activation of the canonical
ate the microbiota. These strategies comprise a struc- WNT–β‑catenin signalling pathway 5. Indeed, mice
tural compartmentalization that minimizes the access in which β‑catenin is constitutively activated in endo­
of bacteria into the underlying lamina propria and their thelial cells are more resistant to Salmonella infection;
systemic dissemination and a specialized immune bar- they show reduced systemic bacterial dissemination, a
rier that ensures intestinal bacteria and food antigens lack of PLVAP expression, which is a marker of vascular
are tolerated (reviewed in REF. 76). Defects in these bar- permeability, and no increase in GVB permeability 5.
riers are responsible for microbial infection, IBDs and
food allergies. Immune regulation at the IEB. Oral tolerance is a state
The physical exclusion of the intestinal microbiota of mucosal and systemic immune unresponsiveness to
from the systemic circulation is achieved by two cellular orally ingested food antigens4. This process requires the
barriers, which include some of the same characteristics uptake of antigens mediated by CX3CR1+ mononuclear
that are present in the BBB and BCSFB of the brain and phagocytic cells, the transfer of these proteins to migra-
in the inner and outer BRB of the eye. Just beneath the tory CD103+ dendritic cells (DCs) via the gap junction
intestinal mucus, the thickness of which differs depend- protein connexin 43 (also known as gap junction‑α1
ing on the location in the intestine77, the first cellular protein) (REF. 84) and the migration of CD103+ DCs
barrier is provided by a monolayer of enterocytes that to the draining lymph nodes85,86, in which they promote
cover the entire mucosa. Similar to the choroid plexus the differentiation of regulatory T cells that have intesti-
epithelium, the IEB is a semipermeable barrier that nal-homing properties87. Alternatively, soluble antigens
limits the passage of molecules and microorganisms can be directly delivered from the intestinal lumen to
through the paracellular route owing to the presence of tolerogenic CD103+ DCs by goblet cells88.
tight junctions and adherens junctions between entero- Epithelial cells are also involved in the process of oral
cytes. Specialized enterocytes, such as goblet cells, which tolerance induction by releasing TGFβ, retinoic acid and
produce mucus, and Paneth cells, which secrete anti- thymic stromal lymphopoietin, which are required for
microbial peptides, provide additional support for the the generation of DCs with a tolerogenic phenotype89,90.
maintenance of intestinal barrier function (reviewed in Food antigens also have access to the intestinal vascula-
REFS 75,78) (FIG. 3a). ture through which they spread systemically 91. Unlike
Lamina propria
The second intestinal barrier — the gut–vascular bar- soluble antigens, commensal microorganisms must be
The layer of mucosal tissue
directly under the mucosal rier (GVB) — is composed of the intestinal endo­thelium, excluded from the blood circulation to avoid the devel-
epithelial cell surface in which which actively blocks haematogenous bacterial dis­ opment of systemic immune activation. In the steady
effector cells for mucosal semination5. Intestinal endothelial cells of the GVB share state, intestinal microorganisms can be found in mes-
immunity reside. many key characteristics with other vascular barriers, enteric lymph nodes (but not in the liver 92) that act as a
Goblet cells
such as specialized intercellular junctional complexes firewall to avoid systemic dissemination via the lymph
Mucus-producing cells found in that reduce the paracellular trafficking of molecules and through the thoracic duct 93.
the epithelial-cell lining of the the expression of different classes of transporters (such as
intestine and lungs. ATP-binding cassette transporters and sugar transport- Regulatory or protective barriers?
ers)5,79. Similar to the structure of the BBB, endothelial On the basis of their structure, cellular components
Paneth cells
Specialized enterocytes that cells in the intestine are closely associated with pericytes and anatomical location (parenchyma or periphery),
are present at the base of the and enteric glial cells (the intestinal counterpart of brain the barriers described above can be grouped into three
crypts in the intestinal astrocytes), which together form the gut–vascular unit 5 main categories: first, protective barriers consisting of
epithelium and that produce (FIG. 3b). The role of these cells in the development and tightly regulated endothelial layers, such as the BBB and
antimicrobial proteins and
peptides, including
maintenance of the GVB in healthy and diseased states the inner BRB; second, more permissive endothelial
phospholipase A2 and has not yet been investigated. However, some studies layers, such as the GVB; and third, immunomodulatory
defensins. suggest that the enteric glial cells have an important role selective gateways consisting of epithelial cells working

6 | ADVANCE ONLINE PUBLICATION www.nature.com/nri


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

b GVB
alone, such as the BCSFB, or working together with an
endothelial barrier, such as the IEB with the GVB or the
Enteric
glial cell outer BRB with the inner BRB.
In the steady state, endothelial barriers are considered
to be protective immunological barriers, as they physi-
cally block leukocyte migration. The BBB endothelium
suppresses parenchymal invasion by leukocytes through
Pericyte
Basement the production of mediators such as IL‑25, which
membrane induces the upregulation of junctional components and
a IEB the downregulation of pro-encephalitogenic cytokines94.
Fenestrated In addition, parenchymal vessels do not express the
Lumen endothelium
ad­­hesion molecule P‑selectin, thereby reducing leuko-
Tight cyte recruitment 95, or express CXC-chemokine ligand
junction 12 on the abluminal side, thereby retaining mono-
Intestinal
nuclear cells in the perivascular space96. By contrast,
microbiota immunosurveillance occurs in the choroid plexus,
where the BCSFB allows the migration of CD4+ effec-
tor memory T cells specific for CNS antigens97. In addi-
tion, and similarly to other epithelial barriers such as
the IEB, the BCSFB can skew immune cells towards
a regulatory phenotype owing to the production of
Mucus TGFβ, vaso­active intestinal peptide98, prostaglandin D2
layer (REF. 99) and IL‑1 receptor antagonist (IL‑1RA)100. Cells
in the choroid plexus, as in the intestine87,101,102, express
Goblet the enzyme indoleamine 2,3‑dioxygenase (IDO), which
cell is responsible for tryptophan catabolism, and retinoic
acid metabolic enzymes, which are involved in the inhi-
Epithelial
bition of T cell activity and the induction of regulatory
cell T cells at the barrier interface10. In the eye, similar prop-
Capillary
Tight junction network erties of immune cell exclusion apply to the inner endo­
thelial BRB, and the ability to skew immune responses is a
CX3CR1+ DC
feature of the outer BRB10.
GSNO For all the barriers described above, the loss of func-
tionality and leukocyte invasion into the correspond-
CD103+ DC
? ing parenchyma can lead to pathological conditions10.
However, it is worth noting that immune cell migra-
Basement
membrane RA Enteric tion into an immune-privileged site is not harmful in all
glia Arteriole cases. Indeed, mice lacking adaptive immune cells show
Treg cell Venule Nerve marked impairments of hippocampal neuro­genesis,
TGFβ TSLP memory and spatial learning abilities103. The T cells
responsible for hippocampal plasticity recognize CNS-
derived autoantigens103, indicating that the BBB may
Lymphatic
vessel be less restrictive than originally thought. Lymphocyte
Lamina propria
patrolling of the CNS is crucial for controlling viral
infections, as illustrated by the failure of the clinical
Figure 3 | The intestinal barriers. In the intestine, epithelial-
Natureand endothelial-based
Reviews | Immunology trial testing the α4β1 integrin (VLA4)‑specific antibody
barriers can be identified. a | The intestinal epithelial barrier (IEB) is the first layer of
defence that prevents the uncontrolled translocation of commensal bacteria from natalizumab in patients with MS104. Natalizumab has
the intestinal lumen into the lamina propria. Goblet cells, which produce the mucus robust beneficial effects on MS disease activity by pre-
layer, and Paneth cells (not depicted), which secrete antimicrobial peptides, give venting extravasation through the BBB of lymphocytes
additional support to the maintenance of mucosal barrier function. A rim of tight expressing α4β1 integrin or α4β7 integrin. However,
junctions seals the epithelial barrier and controls paracellular trafficking. In the by also blocking the recruitment of virus-specific
lamina propria, the mucosal immune system participates in the induction of oral cytolytic cells, treatment with natalizumab has been
tolerance. b | Blood capillaries are found just below the intestinal epithelium. They associated with an increased risk of JC virus infec-
consist of endothelial cells that express junctional proteins to form the gut–vascular tion and the development of progressive multifocal
barrier (GVB). Closely associated with the endothelial cells are enteric glial cells and leukoencephalopathy 104.
pericytes, which together form the gut–vascular unit. Enteric glia cells are involved
Given these examples, it is important to high-
in preserving the integrity of the epithelial and endothelial barriers through the
release of S‑nitrosoglutathione (GSNO). In the healthy state, the GVB controls the light that barrier classification as epithelial or endo­
type of antigens that translocate into the systemic circulation and prohibits the thelial in some cases could be too general, as barrier
entry of the intestinal microbiota. DC, dendritic cell; RA, retinoic acid; TGFβ, systems could work differently depending on their
transforming growth factor‑β; Treg cell, regulatory T cell; TSLP, thymic stromal function and location. For example, size exclusion by
lymphopoietin. the brain and by intestinal vascular barriers is different.

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 7


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Pyramidal neurons The BBB has a size exclusion threshold of 500 Da, pyramidal neurons107. The same disorders can also affect
Type of nerve cells with a whereas the GVB is permeable to molecules as large the BRB in pigs108. Increased permeability of the BRB is
pyramidal-shaped cell body as 4 kDa. This probably reflects the need of the GVB to associated with a leak of plasma components into the
(soma) and two allow the passage of nutrients and antigens for tolerance retina, leading to IgG deposition in the ganglion cell
distinct dendrite trees (the
longer apical and the shorter
induction. In addition, the GVB can afford to be less layer 108. Together, these reports indicate that vascular
basal trees). They are present stringent than the BBB because the first intestinal barrier barriers in different regions of the body are linked and
in most mammalian forebrain is provided by the epithelial layer that directly faces the that they may be disrupted under the same pathological
structures (cerebral cortex, intestinal lumen. conditions, suggesting common mechanisms of
hippocampus and amygdala),
Another degree of complexity is created by the influ- maintenance and protection.
and they are associated with
advanced cognitive functions.
ence of the environment on the barriers — for example, What drives the initial breakdown of vascular barri-
the interplay between the microbiota and the intestinal ers remains to be established, but the observation that
barriers (FIG. 4). Germ-free mice, which lack a normal the IEB is affected in several disorders raises the possi-
intestinal microbiota, have an impaired BBB both in bility that a disturbed epithelial barrier (‘leaky gut’) may
fetal and adult phases owing to a dysregulation of junc- be responsible for vascular barrier breakdown. This is
tional occludin and claudin 5 expression16. However, particularly evident in the intestine, where we demon-
this defect in BBB tight junctions can be repaired strated that pathogens that are capable of disrupting the
when mice are re‑colonized with the microbiota from IEB, such as Salmonella enterica subsp. enterica serovar
mice raised in specific pathogen-free conditions or Typhimurium, can also disrupt the GVB. However, the
are monocolonized with bacteria that produce short- capacity of S. Typhimurium to dismantle the GVB and
chain fatty acids (SCFAs)16. The exact mechanisms by disseminate systemically is an active mechanism that is
which the microbiota affects BBB maturity and function used by the bacteria itself and that depends on its patho­
remain unknown. One possibility is that the intestinal genicity island 2 (REF. 5). Thus, IEB disruption alone is
microbiota influences the IEB and/or the GVB, favour- not sufficient for the microbiota to enter the systemic
ing the translocation of bacteria-derived metabolites105 circulation through the GVB.
into the blood and thus influencing more barriers at A series of reports has shown that patients with
once. Additionally, the microbiota might modulate both autism spectrum disorders (ASDs) experience gastro­
intestinal barrier permeability and brain development intestinal discomfort, which was initially thought
through neurotransmitters (such as serotonin)106. to have a neurobehavioural aetiology rather than a
gastro­e nteric aetiology 109–111. However, subsequent
Barriers and diseases studies have shown that gastrointestinal symptoms in
Neurological disorders.Chronic metabolic disorders individuals with an ASD are associated with mild lev-
associated with nephropathy or retinopathy, such as els of mucosal inflammation112,113. Assessment of intes-
diabetes mellitus or hypercholesterolaemia, can result tinal permeability identified that a high percentage
in an increase in BBB permeability in the cerebral of patients with ASDs (36.7%) and their first-degree
cortex and a progression towards Alzheimer disease, relatives (21.2%) presented with increased permea-
concomitant with deposition of amyloid-β peptide in bility compared with healthy children (4.8%), which
supports the hypothesis that a disturbed IEB and/or
GVB is a distinctive feature of patients with ASDs114.
Gut microbiota
These reports suggest that modifications of the IEB
are linked to dis­orders affecting sites that are far from
Barrier homeostasis the intestine. In this context, both animal and human
studies support a connection between increased intes-
tinal permeability and neurological disorders, includ-
ing ASDs, schizophrenia, Parkinson disease and
GVB and IEB BBB BRB Alzheimer disease. In particular, a post-mortem study
• Tight junction expression • Tight junction expression • Neovascularization has shown that the expression of several genes associ-
modulation modulation • Immune cell recruitment ated with tight junctions is dysregulated in the brains
• Formation of the enteric • Barrier permeability and activation of patients with ASDs and schizophrenia. In particular,
glia network regulation • Establishment of tolerance
• Angiogenesis and vascular • Microglia activation levels of mRNAs encoding claudin 5 and claudin 12 are
remodelling • Modulation of behaviour upregulated in the brain cortex of patients with ASDs
• Modulation of barrier
permeability compared with those of patients with schizophrenia
• Establishment of oral and and healthy controls, whereas claudin 3 expression is
mucosal tolerance upregulated both in patients with schizophrenia and in
those with ASDs115. Expression of the matrix metallo­
Figure 4 | Impact of the microbiota on physiological barrier functions.
Nature ReviewsThe intestinal
| Immunology proteinases MMP2 and MMP9, as well as of trans­
microbiota can affect the physiology and barrier functions in the intestine and in distant
locator protein (TSPO), is also upregulated in patients
organs. In the intestine, it reduces intestinal permeability, induces the establishment of
the gut–vascular unit necessary for correct functioning of the gut–vascular barrier (GVB) with ASDs, whereas tissue-type plasminogen activa-
and enhances mucosal immunity and oral tolerance. At distant sites, such as the tor (tPA) and allograft inflammatory factor 1 (AIF1;
brain and eye, it is involved in modulating vascular barrier permeability and immune also known as IBA1) are downregulated in patients
cell recruitment and activation. BBB, blood–brain barrier; BRB, blood–retinal barrier; with schizophrenia. Together, these data suggest that
IEB, intestinal epithelial barrier. neuro­inflammation (indicated by increased MMP9

8 | ADVANCE ONLINE PUBLICATION www.nature.com/nri


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Dysbiosis and TSPO expression) in patients with ASDs results in barriers share common pathways for their formation
A condition in which the compensatory mechanisms that aim to increase barrier and maintenance, genetic defects or environmental
balance of the bacterial integrity (indicated by increased claudin 5 and claudin triggers affecting one of the pathways involved in bar-
communities that constitute 12 expression at the mRNA level)116. However, protein rier formation may concomitantly affect several barri-
the intestinal microbiota is
altered. Dysbiosis may be a
levels of claudin 12 do not increase in patients with ers in the body. This may explain why diabetes-related
predisposition factor for ASDs, suggesting a failure to increase barrier tight junc- nephropathies and retinopathies are often associated
several diseases. tions. The expression of tight junction proteins is also with BBB disruptions107. A relationship between the
dysregulated in the intestine of patients with ASDs com- intestinal microbiota, IEB permeability and ASDs has
Lewy pathology
pared with healthy controls, which is consistent with the been shown in a mouse model of maternal immune
A pathology characterized by
the presence of abnormal
leaky gut hypothesis115. Similar data have been obtained activation-induced experimental autism118. Changes
α‑synuclein aggregates (bodies) from a mouse model of obesity, in which it was shown in the microbiota due to inflammation during preg-
in the residual neurons of the that anxiety-like behaviour was linked to an impair- nancy were suggested to cause intestinal barrier defects
substantia nigra and in the ment of intestinal barriers, increased expression of in the offspring that are responsible for ASD develop-
enteric nervous system. Lewy
bodies are the histological
neuro­inflammatory markers (such as MMP9, AIF1 and ment. Oral administration of the commensal bacteria
hallmark of Parkinson disease. Toll-like receptors 4 and 2) in the prefrontal cortex and Bacteroides fragilis could correct IEB permeability and
reduced expression of BBB junctional proteins (such as microbiota alterations, leading to reduced symptoms
Microglia claudin 5 and tight junction protein ZO1)117. Together, of ASDs118. Moreover, studies in the context of EAE
Phagocytic cells of myeloid
these results indicate that ASD is often accompanied have demonstrated that the intestinal microbiota can
origin that are involved in the
innate immune response in the
by BBB and IEB defects, linking these two barriers in a influence neurological inflammation119.
central nervous system. gut–brain axis. Given that most of these studies assessed Colonic inflammation has been reported in patients
Microglia are thought to be the the expression of tight junction proteins in the intestine with other neurological disorders such as Parkinson dis-
major brain-resident without separating epithelial cells from endothelial cells, ease, independent of the presence of an enteric Lewy pathol-
macrophages.
it is not clear whether patients with ASDs have a defect ogy120. Interestingly, patients with Parkinson disease show
Autophagy in the GVB as well as in the IEB. α‑synuclein aggregation in neurons of the gastro­intestinal
A specialized process involving What could be the implications of an impaired IEB tract, suggesting that the disease pathology arises in the
the degradative delivery of a or BBB in the development of these disorders? Defects intestine; this pathology does not seem to be a conse-
portion of the cytoplasm or of
in the IEB that may be due to microbiota dysbiosis may quence of inflammation, as patients with IBD did not dis-
damaged organelles to the
lysosome. Internalized
also lead to alterations in the GVB and subsequent play similar aggregation patterns121. Further evidence for
pathogens can also be abnormal translocation of nutrients, microbial prod- the progression of Parkinson disease pathology from the
eliminated by this pathway. ucts or even microorganisms into the systemic circula- intestine to the brain is provided by a recent report show-
tion (FIG. 5). These intestine-derived factors may affect ing active retrograde transport of aggregated α‑synuclein
the BBB. Alternatively, because the different vascular via the vagal nerve (which links to the intestine) to the
dorsal motor neuron122. This begs the question of how
and why α‑synuclein aggregates in the intestine and
Genotype Stress Diet Antibiotics Inflammation whether α‑synuclein aggregation is linked to increased
per­m eability of the IEB. Studies of α‑synuclein-­
overexpressing (ASO) mice, which present a model
of Parkinson disease, confirm a role for the intestinal
Gut microbiota microbiota in the disease123. ASO mice raised in germ-
free conditions show reduced signs of motor neuron
impairment123. It is likely that this effect of the micro­biota
Dysbiosis
occurs during a postnatal phase, as germ-free ASO mice
that are recolonized with a normal micro­biota display
Barrier function impairment features of Parkinson disease, and antibiotic treatment
of adults reduces symptoms of Parkinson disease123. Most
of the Parkinson disease-inducing effects of the micro­
biota can be recapitulated by microbiota-­derived SCFAs.
SCFAs have been shown to be important for the matura-
BBB GVB and IEB BRB tion of brain mononuclear phagocytes, known as micro-
• Anxiety and depression • Metabolic disorders • Type 1 diabetes or glia124. SCFAs may therefore cross the BBB and induce
• Autism spectrum disorders • Autoimmune disorders hypercholesterolaemia- the maturation of microglia, which are then responsi-
• Parkinson disease, (coeliac disease) associated retinopathy
Alzheimer disease and • Inflammatory bowel diseases • Autoimmune uveitis ble for mediating neuroinflammation and α‑synuclein
schizophrenia • Food allergies aggregation. The microbiota or its meta­bolites may
also affect autophagy, thereby impeding the clearance of
Figure 5 | Effect of dysbiosis and barrier failure on disease onset. Many genetic α‑synuclein aggregates123. A similar acceleration of the
and environmental factors affect the composition of the Nature Reviews
intestinal | Immunology
microbiota.
disease was observed when the microbiota from patients
Dysbiosis has been linked to barrier impairments and the development of diseases,
including anxiety and neurological disorders, metabolic disorders and autoimmune with Parkinson disease was transferred into germ-free
diseases. Several studies have reported an association between these diseases and mice, suggesting that dysbiosis leads to disease progres-
increased intestinal permeability that, in turn, may lead to dysregulation of the sion, presumably through the release of SCFAs123. An
blood–brain barrier (BBB) or blood–retinal barrier (BRB). GVB, gut–vascular barrier; important question that arises is whether this dysbiosis
IEB, intestinal epithelial barrier. also promotes increased permeability of the IEB and/or

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 9


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

GVB (maybe through altered expression of as‑yet-un- It would be interesting to determine whether vascular
known receptors) and translocation of SCFAs and other barriers are affected by dietary intervention to reduce
microbiota-derived metabolites into the bloodstream. diabetic complications.
It is unlikely that this effect on the barrier is due to SCFAs, Metabolic disorders are also linked to other dis-
as they have been shown in vitro to increase epithelial eases. Major depressive disorder has comorbidities
integrity rather than increase permeability 125. with type 2 diabetes mellitus, irritable bowel syndrome
An additional mechanism to account for diseases and obesity 137. All these diseases have been associated
mediated by defective vascular barriers may relate with microbiota dysbiosis, reduced physical activity,
to nutrient transport. A recent report has shown that poor diet and a leaky gut (FIG. 5). Microbiota dysbiosis
impaired amino acid transport at the BBB may partic- may lead to the release of metabolites that may affect
ipate in ASD development 126. Mice lacking the solute IEB and GVB permeability, on the one hand, and the
carrier transporter Slc7a5, a neutral amino acid trans- enteric nervous system, on the other, with a direct effect
porter, specifically in endothelial cells have an imbalance on the CNS138. In both cases, the effects propagate to
of amino acid transport into and out of the brain and the whole organism and may lead to comorbidities. One
an accumulation of histidine in the brain that may be interesting hypothesis is that the regulatory function of
involved in causing ASDs126. Histidine may affect the the IEB is lost during dysbiosis and that this induces a
metabolism of brain immune cells. Loss-of-function dysregulation of the GVB. Finally, the microbiota or its
mutations in this amino acid transporter have been metabolic products may access systemic sites, thereby
described in some families with ASDs126. This mutation, contributing to low-grade systemic inflammation and
when present, occurs in every vasculature in the body; leading to several disorders or comorbidities, depending
therefore, a role for amino acid transport may also be on the genetic predisposition of the affected individual.
relevant at other endothelial barriers, in particular in
the intestine, for the translocation of amino acids into Intestinal disorders. IBDs are associated with micro-
the blood. bial dysbiosis139, altered bile acid transformation140 and
submucosal bacterial translocation141. Nearly 30% of
Metabolic and autoimmune disorders. An increase in patients with IBDs have extra-intestinal manifestations
intestinal permeability has been observed in metabolic of the disease, including arthropathies, cutaneous and
and autoimmune disorders and seems to precede overt ocular manifestations and liver and biliary dis­orders142.
disease. In type 1 diabetes (T1D), for example, patients Given that increased epithelial permeability is asso-
with preclinical evidence of the disease also show ciated with metabolic and/or neurodegenerative dis­
increased intestinal permeability, which suggests that orders, it is not clear why extra-intestinal manifestations
diabetes occurs in association with an entero­pathy 127,128. do not occur in all patients with IBDs. It is likely that,
The same is true in spontaneous rodent models of dia- in most of the cases, the GVB functions normally and
betes129. In addition, there is a clear correlation between can therefore completely or partly prevent the trans­
coeliac disease and T1D130. It has been postulated that location of microorganisms or their metabolic products
in the event of higher permeability of the IEB, dietary to systemic organs despite a disrupted IEB. The opposite
antigens may cross the epithelium, become immuno- may be true in patients with coeliac disease, who show
genic and cause autoimmunity 131. Another possible increased serum levels of liver transaminases even when
consequence of increased intestinal permeability is on a gluten-free diet 5. This suggests that liver damage
increased translocation of bacterial products or constit- occurs owing to an impaired GVB even in the absence
uents that may be inflammatory, thereby inducing sys- of damage to the IEB.
temic inflammation. However, what drives the increase
in intestinal permeability is not clear. Dysbiosis has Conclusions and perspectives
been associated with T1D and is character­ized by an In this Review, we describe how endothelial and epi­thelial
under-representation of taxa that produce SCFAs132. barriers often cooperate to establish immune privilege.
Special diets that release SCFAs (either acetate or Endothelial barriers can be more or less stringent depend-
butyrate) have been shown to provide partial pro- ing on the organ that they protect. At body sites that are
tection against T1D, and full protection is conferred classically considered to be immune privileged, barriers
by diets containing both acetate and butyrate132. The tend to exclude immune cells from aberrantly entering
mechanism of action of butyrate involves the induction the organ (such as the BBB in the brain and the BRB in
of regulatory T cells, whereas that of acetate involves the eye). Less stringent barriers, such as the GVB, are not
a reduction of autoreactive T cells133. Although the meant to protect the organ from leuko­cyte migration but
combination of acetate- and butyrate-containing diets rather to avoid the dissemination of unwanted molecules
promotes intestinal integrity 133, protection against T1D to the whole organism. In other words, the GVB protects
is most likely a result of the barrier-­protecting effect the whole body rather than the intestine. Protection of
exerted by acetate134. the intestine is guaranteed by the mucus layer and the
An indication that T1D may be associated with vas- epithelial barrier. The IEB has another important func-
cular impairment comes from the compromised integ- tion in promoting the establishment of tolerance, thereby
rity of the BRB in patients with T1D135, which leads to acting as an immuno­regulatory gate. Hence, the whole
increased frequencies of retinopathies, particularly in body can be considered to be a series of protective barri-
individuals with pregnancy-induced hypertension136. ers: the IEB, which protects the body from the microbiota

10 | ADVANCE ONLINE PUBLICATION www.nature.com/nri


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

but also tolerates it; the GVB, which protects the body with coeliac disease who, even after following a gluten-­
from unwanted molecules that cross the IEB; and the free diet, experience liver damage (as indicated by
other vascular barriers of the body, which protect classical high blood transaminase levels) may benefit from the
immune-privileged organs. These organs have additional administration of drugs aimed at restoring the GVB.
regulatory gates that promote immune tolerance and Similarly, patients with ASDs or ‘high-risk’ individ­
avoid inflammatory responses by leukocytes and thereby uals with increased intestinal permeability may benefit
ensure organ homeostasis and proper development. from treatments aimed at restoring the IEB and GVB
In several diseases, the IEB and possibly the GVB are to both treat and prevent their disorders. In addition,
compromised, allowing leakage of unwanted molecules future research aimed at understanding the mechanisms
into the circulation and perhaps causing damage to the controlling intestinal barriers may help to determine the
other barriers. Knowledge of how barriers are formed mechanisms of barrier formation at other sites and vice
and disrupted may lead to the identification of new versa. Attention should be focused particularly on the
thera­peutic targets to re-establish the epithelial barrier, microbiota and its postbiotics and on the WNT and
the endothelial barrier or both. For example, patients SHH signalling pathways.

1. Medawar, P. B. Immunity to homologous grafted 17. Sharon, G., Sampson, T. R., Geschwind, D. H. & 35. Raab, S. et al. Impaired brain angiogenesis and
skin; the fate of skin homografts transplanted to the Mazmanian, S. K. The central nervous system and the neuronal apoptosis induced by conditional homozygous
brain, to subcutaneous tissue, and to the anterior gut microbiome. Cell 167, 915–932 (2016). inactivation of vascular endothelial growth factor.
chamber of the eye. Br. J. Exp. Pathol. 129, 58–69 18. Amaral, F. A. et al. Commensal microbiota is Thromb. Haemost. 91, 595–605 (2004).
(1948). fundamental for the development of inflammatory 36. Daneman, R. et al. Wnt/ß-catenin signaling is required
2. Iweala, O. I. & Nagler, C. R. Immune privilege in the pain. Proc. Natl Acad. Sci. USA 105, 2193–2197 for CNS, but not non-CNS, angiogenesis.
gut: the establishment and maintenance of non- (2008). Proc. Natl Acad. Sci. USA 106, 641–646 (2009).
responsiveness to dietary antigens and commensal 19. Diaz Heijtz, R. et al. Normal gut microbiota modulates 37. Liebner, S. et al. Wnt/ß-catenin signaling controls
flora. Immunol. Rev. 213, 82–100 (2006). brain development and behavior. Proc. Natl Acad. Sci. development of the blood-brain barrier. J. Cell Biol.
3. Chistiakov, D. A., Bobryshev, Y. V., Kozarov, E., USA 1 08, 3047–3052 (2011). 183, 409–417 (2008).
Sobenin, I. A. & Orekhov, A. N. Intestinal mucosal This study provides evidence that microbial 38. Kuhnert, F. et al. Essential regulation of CNS
tolerance and impact of gut microbiota to mucosal colonization regulates signalling mechanisms, angiogenesis by the orphan G protein-coupled
tolerance. Front. Microbiol. 6, 781 (2015). neurotransmitter turnover and synaptic-related receptor GPR124. Science 330, 985–989 (2010).
4. Pabst, O. & Mowat, A. M. Oral tolerance to food protein production to affect motor control and 39. Anderson, K. D. et al. Angiogenic sprouting into neural
protein. Mucosal Immunol. 5, 232–239 (2012). anxiety behaviour. tissue requires Gpr124, an orphan G protein-coupled
5. Spadoni, I. et al. A gut-vascular barrier controls the 20. Al‑Asmakh, M. et al. The gut microbiota and receptor. Proc. Natl Acad. Sci. USA 108, 2807–2812
systemic dissemination of bacteria. Science 350, developmental programming of the testis in mice. (2011).
830–834 (2015). PLoS ONE 9, e103809 (2014). 40. Cullen, M. et al. GPR124, an orphan G protein-
This study demonstrates the existence of the GVB 21. Andriessen, E. M. et al. Gut microbiota influences coupled receptor, is required for CNS-specific
and shows that endothelial cells control the pathological angiogenesis in obesity-driven choroidal vascularization and establishment of the blood-brain
passage of antigens into the bloodstream and neovascularization. EMBO Mol. Med. 8, e201606531 barrier. Proc. Natl Acad. Sci. USA 108, 5759–5764
prohibit entry of the microbiota. (2016). (2011).
6. Forrester, J. V., Xu, H., Lambe, T. & Cornall, R. Immune 22. Engelhardt, B. & Ransohoff, R. M. Capture, crawl, 41. Hellström, M. et al. Lack of pericytes leads to
privilege or privileged immunity? Mucosal Immunol. cross: The T cell code to breach the blood-brain endothelial hyperplasia and abnormal vascular
1, 372–381 (2008). barriers. Trends Immunol. 33, 579–589 (2012). morphogenesis. J. Cell Biol. 153, 543–553 (2001).
7. Joyce, J. A. & Fearon, D. T. T cell exclusion, immune 23. Nicholas, M. K., Antel, J. P., Stefansson, K. & 42. Daneman, R., Zhou, L., Kebede, A. A. & Barres, B. A.
privilege, and the tumor microenvironment. Arnason, B. G. Rejection of fetal neocortical neural Pericytes are required for blood-brain barrier integrity
Science 348, 74–80 (2015). transplants by H-2 incompatible mice. J. Immunol. during embryogenesis. Nature 468, 562–566 (2010).
8. Carson, M. J., Doose, J. M., Melchior, B., 139, 2275–2283 (1987). 43. Armulik, A. et al. Pericytes regulate the blood-brain
Schmid, C. D. & Ploix, C. C. CNS immune privilege: 24. Mason, D. W. et al. The fate of allogeneic and xenogeneic barrier. Nature 468, 557–561 (2010).
Hiding in plain sight. Immunol. Rev. 213, 48–65 neuronal tissue transplanted into the third ventricle of 44. Bell, R. D. et al. Pericytes control key neurovascular
(2006). rodents. Neuroscience 19, 685–694 (1986). functions and neuronal phenotype in the adult brain
9. Louveau, A., Harris, T. H. & Kipnis, J. Revisiting the 25. Obermeier, B., Daneman, R. & Ransohoff, R. M. and during brain aging. Neuron 68, 409–427 (2010).
mechanisms of CNS immune privilege. Development, maintenance and disruption of the The studies in references 42–44 demonstrate a
Trends Immunol. 36, 569–577 (2015). blood-brain barrier. Nat. Med. 19, 1584–1596 clear role for pericytes in different phases of BBB
10. Shechter, R., London, A. & Schwartz, M. Orchestrated (2013). development and during ageing.
leukocyte recruitment to immune-privileged sites: 26. Alvarez, J. I. et al. The Hedgehog pathway promotes 45. Lee, S.‑W. et al. SSeCKS regulates angiogenesis and
absolute barriers versus educational gates. blood-brain barrier integrity and CNS immune tight junction formation in blood-brain barrier.
Nat. Rev. Immunol. 13, 206–218 (2013). quiescence. Science 334, 1727–1731 (2011). Nat. Med. 9, 900–906 (2003).
This article summarizes literature on barriers at 27. Greter, M. et al. Dendritic cells permit immune 46. Bell, R. D. et al. Apolipoprotein E controls
immune-privileged sites. invasion of the CNS in an animal model of multiple cerebrovascular integrity via cyclophilin A.
11. Goto, Y. & Kiyono, H. Epithelial barrier: an interface sclerosis. Nat. Med. 11, 328–334 (2005). Nature 485, 512–516 (2012).
for the cross-communication between gut flora and 28. Owens, T., Bechmann, I. & Engelhardt, B. Perivascular 47. Methia, N. et al. ApoE deficiency compromises the
immune system. Immunol. Rev. 245, 147–163 spaces and the two steps to neuroinflammation. blood brain barrier especially after injury. Mol. Med.
(2012). J. Neuropathol. Exp. Neurol. 67, 1113–1121 (2008). 7, 810–815 (2001).
12. Engelhardt, B., Vajkoczy, P. & Weller, R. O. The movers 29. Hickey, W. F. & Kimura, H. Perivascular microglial cells 48. Hafezi-Moghadam, A., Thomas, K. L. & Wagner, D. D.
and shapers in immune privilege of the CNS. of the CNS are bone marrow-derived and present ApoE deficiency leads to a progressive age-dependent
Nat. Immunol. 18, 123–131 (2017). antigen in vivo. Science 239, 290–292 (1988). blood-brain barrier leakage. AJP Cell Physiol. 292,
13. Stappenbeck, T. S., Hooper, L. V. & Gordon, J. I. 30. Williams, K., Alvarez, X. & Lackner, A. A. Central C1256–C1262 (2006).
Developmental regulation of intestinal angiogene sis nervous system perivascular cells are 49. Mizee, M. R. et al. Retinoic acid induces blood-brain
by indigenous microbes via Paneth cells. Proc. Natl immunoregulatory cells that connect the CNS with the barrier development. J. Neurosci. 33, 1660–1671
Acad. Sci. USA 99, 15451–15455 (2002). peripheral immune system. Glia 36, 156–164 (2001). (2013).
14. Reinhardt, C. et al. Tissue factor and PAR1 promote 31. Harris, M. G. et al. Immune privilege of the CNS is not 50. Liddelow, S. A. Development of the choroid plexus and
microbiota-induced intestinal vascular remodelling. the consequence of limited antigen sampling. Sci. Rep. blood-CSF barrier. Front. Neurosci. 9, 32 (2015).
Nature 483, 627–631 (2012). 4, 4422 (2014). 51. Lun, M. P., Monuki, E. S. & Lehtinen, M. K.
15. Hooper, L. V. et al. Molecular analysis of commensal 32. Stewart, P. A. & Wiley, M. J. Developing nervous tissue Development and functions of the choroid plexus–
host-microbial relationships in the intestine. induces formation of blood-brain barrier cerebrospinal fluid system. Nat. Rev. Neurosci. 16,
Science 291, 881–884 (2001). characteristics in invading endothelial cells: a study 445–457 (2015).
16. Braniste, V. et al. The gut microbiota influences blood- using quail-chick transplantation chimeras. Dev. Biol. 52. Johansson, P. A. The choroid plexuses and their
brain barrier permeability in mice. Sci. Transl. Med. 6, 84, 183–192 (1981). impact on developmental neurogenesis.
263ra158 (2014). 33. Janzer, R. C. & Raff, M. C. Astrocytes induce blood- Front. Neurosci. 8, 340 (2014).
This study demonstrates that the intestinal brain barrier properties in endothelial cells. 53. Steinemann, A., Galm, I., Chip, S., Nitsch, C. &
microbiota can shape BBB properties. It shows that Nature 325, 253–257 (1987). Maly, I. P. Claudin‑1, -2 and -3 are selectively
mice lacking a normal intestinal microbiota have 34. Stenman, J. M. et al. Canonical Wnt signaling expressed in the epithelia of the choroid plexus of the
increased BBB permeability compared with regulates organ-specific assembly and differentiation mouse from early development and into adulthood
pathogen-free mice and that recolonization of of CNS vasculature. Science 322, 1247–1250 while claudin‑5 is restricted to endothelial cells.
germ-free adult mice restores BBB functionality. (2008). Front. Neuroanat. 10, 16 (2016).

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 11


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

54. Kratzer, I. et al. Complexity and developmental 77. Pelaseyed, T. et al. The mucus and mucins of the patients. Trans. R. Soc. Trop. Med. Hyg. 84, 795–799
changes in the expression pattern of claudins at the goblet cells and enterocytes provide the first defense (1990).
blood-CSF barrier. Histochem. Cell Biol. 138, line of the gastrointestinal tract and interact with the 100. Tarkowski, E., Liljeroth, A. M., Nilsson, A., Minthon, L.
861–879 (2012). immune system. Immunol. Rev. 260, 8–20 (2014). & Blennow, K. Decreased levels of intrathecal
55. Wolburg, H., Wolburg-Buchholz, K., Liebner, S. & 78. Daneman, R. & Rescigno, M. The gut immune barrier interleukin 1 receptor antagonist in Alzheimer’s
Engelhardt, B. Claudin‑1, claudin‑2 and claudin‑11 are and the blood-brain barrier: are they so different? disease. Dement. Geriatr. Cogn. Disord. 12, 314–317
present in tight junctions of choroid plexus epithelium Immunity 31, 722–735 (2009). (2001).
of the mouse. Neurosci. Lett. 307, 77–80 (2001). 79. Spadoni, I., Pietrelli, A., Pesole, G. & Rescigno, M. 101. Coombes, J. L. et al. A functionally specialized
56. Lun, M. P., Monuki, E. S. & Lehtinen, M. K. Gene expression profile of endothelial cells during population of mucosal CD103+ DCs induces Foxp3+
Development and functions of the choroid plexus– perturbation of the gut vascular barrier. Gut Microbes regulatory T cells via a TGF-ß and retinoic acid-
cerebrospinal fluid system. Nat Rev Neurosci. 16, 7, 1–9 (2016). dependent mechanism. J. Exp. Med. 204,
445–457 (2015). 80. Bush, T. G. et al. Fulminant jejuno-ileitis following 1757–1764 (2007).
57. Caspi, R. R. Ocular autoimmunity: the price of ablation of enteric glia in adult transgenic mice. 102. Matteoli, G. et al. Gut CD103+ dendritic cells express
privilege? Immunol. Rev. 213, 23–35 (2006). Cell 93, 189–201 (1998). indoleamine 2,3‑dioxygenase which influences T
58. Zhou, L. et al. The Schlemm’s canal is a VEGF‑C/ 81. Cornet, A. et al. Enterocolitis induced by autoimmune regulatory/T effector cell balance and oral tolerance
VEGFR‑3–responsive lymphatic-like vessel. targeting of enteric glial cells: a possible mechanism in induction. Gut 59, 595–604 (2010).
Mol. Vis. 12, 639–643 (2014). Crohn’s disease? Proc. Natl Acad. Sci. USA 98, 103. Ziv, Y. et al. Immune cells contribute to the
59. Kaur, C., Foulds, W. S. & Ling, E. A. Blood–retinal 13306–13311 (2001). maintenance of neurogenesis and spatial learning
barrier in hypoxic ischaemic conditions: basic 82. Savidge, T. C. et al. Enteric glia regulate intestinal abilities in adulthood. Nat. Neurosci. 9, 268–275
concepts, clinical features and management. barrier function and inflammation via release of (2006).
Prog. Retin. Eye Res. 27, 622–647 (2008). S‑nitrosoglutathione. Gastroenterology 132, 104. Antoniol, C. & Stankoff, B. Immunological markers for
60. Kim, J. H., Kim, J. H., Yu, Y. S., Kim, D. H. & Kim, K. W. 1344–1358 (2007). PML prediction in MS patients treated with
Recruitment of pericytes and astrocytes is closely 83. Kabouridis, P. S. et al. Microbiota controls the natalizumab. Front. Immunol. 6, 668 (2015).
related to the formation of tight junction in developing homeostasis of glial cells in the gut lamina propria. 105. Tsilingiri, K. & Rescigno, M. Postbiotics: what else?
retinal vessels. J. Neurosci. Res. 87, 653–659 Neuron 85, 289–295 (2015). Benef. Microbes 4, 101–107 (2013).
(2009). 84. Mazzini, E., Massimiliano, L., Penna, G. & 106. Kelly, J. R. et al. Breaking down the barriers: the gut
61. Yao, H. et al. The development of blood-retinal Rescigno, M. Oral tolerance can be established via microbiome, intestinal permeability and stress-related
barrier during the interaction of astrocytes with gap junction transfer of fed antigens from CX3CR1+ psychiatric disorders. Front. Cell. Neurosci. 9, 392
vascular wall cells. Neural Regen. Res. 9, macrophages to CD103+ dendritic cells. Immunity 40, (2015).
1047–1054 (2014). 248–261 (2014). 107. Acharya, N. K. et al. Diabetes and
62. Tout, S., Chan-Ling, T., Holländer, H. & Stone, J. The 85. Bogunovic, M. et al. Origin of the lamina propria hypercholesterolemia increase blood-brain barrier
role of müller cells in the formation of the blood-retinal dendritic cell network. Immunity 31, 513–525 permeability and brain amyloid deposition: beneficial
barrier. Neuroscience 55, 291–301 (1993). (2009). effects of the LpPLA2 inhibitor darapladib.
63. Shen, W. et al. Conditional muller cell ablation causes 86. Schulz, O. et al. Intestinal CD103+, but not CX3CR1+, J. Alzheimers Dis. 35, 179–198 (2013).
independent neuronal and vascular pathologies in a antigen sampling cells migrate in lymph and serve 108. Acharya, N. K. et al. Retinal pathology is associated
novel transgenic model. J. Neurosci. 32, classical dendritic cell functions. J. Exp. Med. 206, with increased blood-retina barrier permeability in a
15715–15727 (2012). 3101–3114 (2009). diabetic and hypercholesterolaemic pig model:
64. Chow, B. W. & Gu, C. Gradual suppression of 87. Sun, C. M. et al. Small intestine lamina propria Beneficial effects of the LpPLA2 inhibitor Darapladib.
transcytosis governs functional blood-retinal barrier dendritic cells promote de novo generation of Foxp3 T Diabetes Vasc. Dis. Res. 14, 200–213 (2017).
formation. Neuron 93, 1325–1333.e3 (2017). reg cells via retinoic acid. J. Exp. Med. 204, 109. Ibrahim, S. H., Voigt, R. G., Katusic, S. K.,
65. Ogura, S. et al. Sustained inflammation after pericyte 1775–1785 (2007). Weaver, A. L. & Barbaresi, W. J. Incidence of
depletion induces irreversible blood-retina barrier 88. McDole, J. R. et al. Goblet cells deliver luminal antigen gastrointestinal symptoms in children with autism: a
breakdown. JCI Insight 2, e90905 (2017). to CD103+ dendritic cells in the small intestine. population-based study. Pediatrics 124, 680–686
66. Simò, R., Villarroel, M., Corraliza, L., Hernàndez, C. & Nature 483, 345–349 (2012). (2009).
Garcia-Ramìrez, M. The retinal pigment epithelium: 89. Iliev, I. D., Mileti, E., Matteoli, G., Chieppa, M. & 110. Klukowski, M., Wasilewska, J. & Lebensztejn, D. Sleep
something more than a constituent of the blood- Rescigno, M. Intestinal epithelial cells promote colitis- and gastrointestinal disturbances in autism spectrum
retinal barrier — implications for the pathogenesis of protective regulatory T‑cell differentiation through disorder in children. Dev. Period Med. 19, 157–161
diabetic retinopathy. BioMed Res. 2010, 1–15 dendritic cell conditioning. Mucosal Immunol. 2, (2015).
(2010). 340–350 (2009). 111. Chaidez, V., Hansen, R. L. & Hertz-Picciotto, I.
67. Rizzolo, L. J. Barrier properties of cultured retinal 90. Iliev, I. D. et al. Human intestinal epithelial cells Gastrointestinal problems in children with autism,
pigment epithelium. Exp. Eye Res. 126, 16–26 promote the differentiation of tolerogenic dendritic developmental delays or typical development.
(2014). cells. Gut 58, 1481–1489 (2009). J. Autism Dev. Disord. 44, 1117–1127 (2014).
68. Sugita, S., Futagami, Y., Smith, S. B., Naggar, H. & 91. Goubier, A. et al. Plasmacytoid dendritic cells mediate 112. Horvath, K. & Perman, J. Autistic disorder and
Mochizuki, M. Retinal and ciliary body pigment oral tolerance. Immunity 29, 464–475 (2008). gastrointestinal disease. Curr. Opin. Pediatr. 14,
epithelium suppress activation of T lymphocytes via 92. Balmer, M. L. et al. The liver may act as a firewall 583–587 (2002).
transforming growth factor beta. Exp. Eye Res. 83, mediating mutualism between the host and its gut 113. Kushak, R. I. et al. Evaluation of intestinal function in
1459–1471 (2006). commensal microbiota. Sci. Transl. Med. 6, 237ra66 children with autism and gastrointestinal symptoms.
69. Fang, Y., Yu, S., Ellis, J. S., Sharav, T. & Braley- (2014). J. Pediatr. Gastroenterol. Nutr. 62, 687–691 (2016).
mullen, H. Comparison of sensitivity of Th1, Th2, This study demonstrates that the liver acts as a 114. de Magistris, L. et al. Alterations of the intestinal
and Th17 cells to Fas-mediated apoptosis. J. Leukoc. vascular firewall that impedes intestinal bacteria barrier in patients with autism spectrum disorders and
Biol. 87, 1019–1028 (2010). from entering the bloodstream during intestinal in their first-degree relatives. J. Pediatr. Gastroenterol.
70. Sohn, J. H., Kaplan, H. J., Suk, H. J., Bora, P. S. & pathology, spreading systemically and activating Nutr. 51, 418–424 (2010).
Bora, N. S. Chronic low level complement activation non-mucosal immune responses. 115. Fiorentino, M. et al. Blood–brain barrier and
within the eye is controlled by intraocular complement 93. Macpherson, A. J. & Smith, K. Mesenteric lymph intestinal epithelial barrier alterations in autism
regulatory proteins. Investig. Ophthalmol. Vis. Sci. 41, nodes at the center of immune anatomy. J. Exp. Med. spectrum disorders. Mol. Autism 7, 49 (2016).
3492–3502 (2000). 203, 497–500 (2006). 116. Nitta, T. et al. Size-selective loosening of the blood-
71. Taylor, A. A review of the influence of aqueous humor 94. Sonobe, Y. et al. Interleukin‑25 expressed by brain brain barrier in claudin‑5‑deficient mice. J. Cell. Biol.
on immunity. Ocul. Immunol. Inflamm. 11, 231–241 capillary endothelial cells maintains blood-brain 161, 653–660 (2003).
(2003). barrier function in a protein kinase C‑dependent 117. Bruce-Keller, A. J. et al. Obese-type gut microbiota
72. Cousins, S. W., McCabe, M. M., Danielpour, D. & manner. J. Biol. Chem. 284, 31834–31842 (2009). induce neurobehavioral changes in the absence of
Streilein, J. W. Identification of transforming growth 95. Barkalow, F. J., Goodman, M. J., Gerritsen, M. E. & obesity. Biol. Psychiatry 77, 607–615 (2015).
factor-beta as an immunosuppressive factor in Mayadas, T. N. Brain endothelium lack one of two 118. Hsiao, E. Y. et al. Microbiota modulate behavioral and
aqueous humor. Investig. Ophthalmol. Vis. Sci. 32, pathways of P‑selectin-mediated neutrophil adhesion. physiological abnormalities associated with
2201–2211 (1991). Blood 88, 4585–4593 (1996). neurodevelopmental disorders. Cell 155, 1451–1463
73. Wilbanks, G. A. & Streilein, J. W. Characterization of 96. McCandless, E. E., Wang, Q., Woerner, B. M., (2013).
suppressor cells in anterior chamber-associated Harper, J. M. & Klein, R. S. CXCL12 limits 119. Lee, Y. K., Menezes, J. S., Umesaki, Y. &
immune deviation (ACAID) induced by soluble antigen. inflammation by localizing mononuclear infiltrates to Mazmanian, S. K. Proinflammatory T‑cell responses to
Evidence of two functionally and phenotypically the perivascular space during experimental gut microbiota promote experimental autoimmune
distinct T‑suppressor cell populations. Immunology 71, autoimmune encephalomyelitis. J. Immunol. 177, encephalomyelitis. Proc. Natl Acad. Sci. USA 108,
383–389 (1990). 8053–8064 (2006). 4615–4622 (2011).
74. Niederkorn, J. Y. See no evil, hear no evil, do no evil: 97. Kivisakk, P. et al. Human cerebrospinal fluid central This work demonstrates that intestinal bacteria
the lessons of immune privilege. Nat. Immunol. 7, memory CD4+ T cells: evidence for trafficking through influence inflammatory immune responses in the
354–359 (2006). choroid plexus and meninges via P‑selectin. Proc. Natl CNS, favouring EAE development.
An exhaustive review of the processes contributing Acad. Sci. USA 100, 8389–8394 (2003). 120. Devos, D. et al. Colonic inflammation in Parkinson’s
to the immune-privileged status of the eye. 98. Taylor, A. W. & Streilein, J. W. Inhibition of antigen- disease. Neurobiol. Dis. 50, 42–48 (2013).
75. Ley, R. E., Peterson, D. A. & Gordon, J. I. Ecological stimulated effector T cells by human cerebrospinal 121. Shannon, K. M. et al. α-Synuclein in colonic
and evolutionary forces shaping microbial diversity in fluid. Neuroimmunomodulation 3, 112–118 (1996). submucosa in early untreated Parkinson’s disease.
the human intestine. Cell 124, 837–848 (2006). 99. Pentreath, V. W., Rees, K., Owolabi, O. A., Philip, K. A. Mov. Disord. 27, 709–715 (2012).
76. Mowat, A. M. Anatomical basis of tolerance and & Doua, F. The somnogenic T lymphocyte suppressor 122. Holmqvist, S. et al. Direct evidence of Parkinson
immunity to intestinal antigens. Nat. Rev. Immunol. 3, prostaglandin D2 is selectively elevated in pathology spread from the gastrointestinal tract to the
331–341 (2003). cerebrospinal fluid of advanced sleeping sickness brain in rats. Acta Neuropathol. 128, 805–820 (2014).

12 | ADVANCE ONLINE PUBLICATION www.nature.com/nri


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

123. Sampson, T. R. et al. Gut microbiota regulate motor 131. Ménard, S., Cerf-Bensussan, N. & Heyman, M. 140. Duboc, H. et al. Connecting dysbiosis, bile-acid
deficits and neuroinflammation in a model of Multiple facets of intestinal permeability and epithelial dysmetabolism and gut inflammation in inflammatory
parkinson’s disease. Cell 167, 1469–1480 (2016). handling of dietary antigens. Mucosal Immunol. 3, bowel diseases. Gut 62, 531–539 (2013).
This study shows the functional role of the 247–259 (2010). This work shows that IBD-associated dysbiosis leads
intestinal microbiota in promoting α‑synucle- 132. De Goffau, M. C. et al. Fecal microbiota composition to modifications in the bile acid pool and consequently
in-mediated brain pathology and motor deficits differs between children with b‑cell autoimmunity and amplifies chronic inflammation processes.
in a mouse model of Parkinson disease. those without. Diabetes 62, 1238–1244 (2013). 141. Chiodini, R. J., Dowd, S. E., Galandiuk, S., Davis, B. &
Interestingly, transplantation of faecal 133. Mariño, E. et al. Gut microbial metabolites limit the Glassing, A. The predominant site of bacterial
microorganisms from patients with Parkinson frequency of autoimmune T cells and protect against translocation across the intestinal mucosal barrier
disease into mice is shown to be sufficient to type 1 diabetes. Nat. Immunol. 18, 552–562 occurs at the advancing disease margin in Crohn’s
impair motor functions. (2017). disease. Microbiol 162, 1608–1619 (2016).
124. Erny, D. et al. Host microbiota constantly control 134. Fukuda, S. et al. Bifidobacteria can protect from 142. Ott, C. & Scholmerich, J. Extraintestinal
maturation and function of microglia in the CNS. enteropathogenic infection through production of manifestations and complications in IBD. Nat. Rev.
Nat. Neurosci. 18, 965–977 (2015). acetate. Nature 469, 543–547 (2011). Gastroenterol. Hepatol. 10, 585–595 (2013).
125. Mariadason, J. M., Barkla, D. H. & Gibson, P. R. Effect 135. Mosier, M. A., Lopez, K. H., Noorbaksh, K. R. &
of short-chain fatty acids on paracellular permeability Charles, M. A. Early retinal and renal abnormalities in Acknowledgements
in Caco‑2 intestinal epithelium model. Am. J. Physiol. diabetes. J. Diabetes Compl. 11, 218–224 (1997). This work is supported by the European Council of Research
272, G705–G712 (1997). 136. Gordin, D. et al. Pre-eclampsia and pregnancy-induced (Consolidator grant HomeoGUT, 615735), the Italian
126. Tărlungeanu, D. C. et al. Impaired amino acid hypertension are associated with severe diabetic Association for Cancer Research (AIRC) and the Italian
transport at the blood brain barrier is a cause of retinopathy in type 1 diabetes later in life. Acta Ministry of Health (Ricerca finalizzata). G.F. is supported by
autism spectrum disorder. Cell 167, 1481–1494 Diabetol. 50, 781–787 (2013). the Italian Ministry of Health (grant GR‑2013‑02359806)
(2016). 137. Slyepchenko, A. et al. Intestinal dysbiosis, gut and by Fondazione Cariplo (grant 2016–0472).
127. Bosi, E. et al. Increased intestinal permeability hyperpermeability and bacterial translocation:
precedes clinical onset of type 1 diabetes. missing links between depression, obesity and Author contributions
Diabetologia 49, 2824–2827 (2006). type 2 diabetes? Curr. Pharm. Des. 22, 6087–6106 I.S. and M.R. wrote the article and reviewed and edited the
128. Sapone, A. et al. Zonulin upregulation is associated (2016). manuscript before submission. G.F. contributed to research-
with increased gut permeability in subjects with type 1 138. Slyepchenko, A. et al. Gut microbiota, bacterial ing data for the article and reviewed and edited the manu-
diabetes and their relatives. Diabetes 55, translocation, and interactions with diet: script before submission.
1443–1449 (2006). pathophysiological links between major depressive
129. Daft, J. G. & Lorenz, R. G. Role of the gastrointestinal disorder and non-communicable medical Competing interests statement
ecosystem in the development of type 1 diabetes. comorbidities. Psychother Psychosom. 8686, The authors declare no competing interests.
Pediatr. Diabetes 16, 407–418 (2015). 31–4631 (2017).
130. Schuppan, D. & Hahn, E. G. Celiac disease and its link 139. Gevers, D. et al. The treatment-naive microbiome in Publisher’s note
to type 1 diabetes mellitus. J. Pediatr. Endocrinol. new-onset Crohn’s disease. Cell Host Microbe 15, Springer Nature remains neutral with regard to jurisdictional
Metab. 14 (Suppl. 1), 597–605 (2001). 382–392 (2014). claims in published maps and institutional affiliations.

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 13


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

You might also like