ArticlePDF AvailableLiterature Review

The Mechanism of Facultative Intracellular Parasitism of Brucella

Authors:

Abstract and Figures

Brucellosis is a highly prevalent zoonotic disease characterized by abortion and reproductive dysfunction in pregnant animals. Although the mortality rate of Brucellosis is low, it is harmful to human health, and also seriously affects the development of animal husbandry, tourism and international trade. Brucellosis is caused by Brucella, which is a facultative intracellular parasitic bacteria. It mainly forms Brucella-containing vacuoles (BCV) in the host cell to avoid the combination with lysosome (Lys), so as to avoid the elimination of it by the host immune system. Brucella not only has the ability to resist the phagocytic bactericidal effect, but also can make the host cells form a microenvironment which is conducive to its survival, reproduction and replication, and survive in the host cells for a long time, which eventually leads to the formation of chronic persistent infection. Brucella can proliferate and replicate in cells, evade host immune response and induce persistent infection, which are difficult problems in the treatment and prevention of Brucellosis. Therefore, the paper provides a preliminary overview of the facultative intracellular parasitic and immune escape mechanisms of Brucella, which provides a theoretical basis for the later study on the pathogenesis of Brucella.
Content may be subject to copyright.
International Journal of
Molecular Sciences
Review
The Mechanism of Facultative Intracellular Parasitism
of Brucella
Hanwei Jiao 1,2,3,*, Zhixiong Zhou 2, Bowen Li 2, Yu Xiao 2, Mengjuan Li 2, Hui Zeng 2, Xiaoyi Guo 2
and Guojing Gu 2


Citation: Jiao, H.; Zhou, Z.; Li, B.;
Xiao, Y.; Li, M.; Zeng, H.; Guo, X.; Gu,
G. The Mechanism of Facultative
Intracellular Parasitism of Brucella.
Int. J. Mol. Sci. 2021,22, 3673.
https://doi.org/10.3390/ijms22073673
Academic Editor: Rustam I. Aminov
Received: 24 February 2021
Accepted: 30 March 2021
Published: 1 April 2021
Publisher’s Note: MDPI stays neutral
with regard to jurisdictional claims in
published maps and institutional affil-
iations.
Copyright: © 2021 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
1Immunology Research Center, Medical Research Institute, Southwest University, Chongqing 402460, China
2College of Veterinary Medicine, Southwest University, Chongqing 402460, China;
zzx449090405@email.swu.edu.cn (Z.Z.); libowenswu@email.swu.edu.cn (B.L.);
xiaoyulike@email.swu.edu.cn (Y.X.); lmj123@email.swu.edu.cn (M.L.); zenghui1234@email.swu.edu.cn (H.Z.);
greataguo4@email.swu.edu.cn (X.G.); ggj19970819@email.swu.edu.cn (G.G.)
3Veterinary Scientific Engineering Research Center, Chongqing 402460, China
*Correspondence: jiaohanwei@swu.edu.cn
Abstract:
Brucellosis is a highly prevalent zoonotic disease characterized by abortion and repro-
ductive dysfunction in pregnant animals. Although the mortality rate of Brucellosis is low, it is
harmful to human health, and also seriously affects the development of animal husbandry, tourism
and international trade. Brucellosis is caused by Brucella, which is a facultative intracellular parasitic
bacteria. It mainly forms Brucella-containing vacuoles (BCV) in the host cell to avoid the combination
with lysosome (Lys), so as to avoid the elimination of it by the host immune system. Brucella not
only has the ability to resist the phagocytic bactericidal effect, but also can make the host cells form a
microenvironment which is conducive to its survival, reproduction and replication, and survive in
the host cells for a long time, which eventually leads to the formation of chronic persistent infection.
Brucella can proliferate and replicate in cells, evade host immune response and induce persistent
infection, which are difficult problems in the treatment and prevention of Brucellosis. Therefore, the
paper provides a preliminary overview of the facultative intracellular parasitic and immune escape
mechanisms of Brucella, which provides a theoretical basis for the later study on the pathogenesis
of Brucella.
Keywords:
Brucella; facultative intracellular parasitism; immune escape; persistent infection; re-
search progress
1. Introduction
Brucellosis is a zoonotic systemic chronic infectious disease caused by Brucella [
1
]. Bru-
cellosis is often called “Mediterranean fever” or “Malta fever”. The main clinical features
of the disease are high fever, enlargement of liver and spleen, joint pain, inflammation
of reproductive organs and fetal membranes, infertility and localized lesions of various
tissues [
2
,
3
]. Brucellosis is widely distributed around the world; only a few countries
in Northern Europe and Central Europe, as well as Canada, Japan, Australia and New
Zealand, have eliminated Brucellosis. The Mediterranean area, Asia and central and South
America are the high incidence areas of Brucellosis. Indeed, Brucellosis has been described
as being the most common zoonotic disease worldwide with more than 500,000 new human
cases annually [1].
Brucella is a Gram-negative facultative intracellular parasitic bacteria—it has no spores,
flagella and capsules [
4
]. In 1886, David Bruce first identified and isolated Brucella from
the spleens of soldiers who died of “Maltese fever” [
5
]. In 1985, the WHO Brucellosis
expert committee divided Brucella spp. into six species according to the differences of
infected animals and antigenicity. Brucella melitensis (B.melitensis) is the most common
and virulent bacteria, followed by Brucella abortus (B.abortus). Brucella mainly infects
Int. J. Mol. Sci. 2021,22, 3673. https://doi.org/10.3390/ijms22073673 https://www.mdpi.com/journal/ijms
Int. J. Mol. Sci. 2021,22, 3673 2 of 13
ruminants and causes abortion and infertility in pregnant animals [
6
]. The Brucella cell
wall consists of an inner and an outer membrane. The outer membrane of Brucella consists
of lipopolysaccharide (LPS), outer membrane protein (OMP) and phospholipid layer.
Lipid A, core polysaccharide and O antigen constitute the LPS of Brucella, which is an
important virulence factor of Brucella and an important antigen that mediates the immune
production of the animal body [
7
]. Brucella mainly invades macrophages and trophoblast
cells, parasitizes in host cells through specific molecular mechanism, affects the apoptosis
of host cells, thus mediates the autophagy of host cells, and creates favorable conditions
for its survival and reproduction in the host cells. After Brucella invades the host cells, it
mainly causes chronic infection by avoiding the host immune-reaction system [
8
], but the
molecular mechanism of Brucella facultative intracellular parasitism has not been clear.
Therefore, through an overview of the mechanism of Brucella facultative intracellular
parasitism, this article provides a theoretical basis for the later excavation of the pathogenic
mechanism of Brucella and related research.
2. Intracellular Life Cycle of Brucella
Brucella has been initially described as a facultative intracellular parasitic bacteria
able to replicate in professional phagocytes such as macrophages, dendritic cells (DC) and
granulocytes as well as nonprofessional phagocytes, including epithelial, fibroblastic and
trophoblastic cells [
9
]. Brucella interacts with the cell membrane of macrophages through
lipid rafts and enters the host cells to form Brucella-containing vacuoles (BCV) surrounded
by phagocytic vesicles [
10
]. From 8 to 12 h after Brucella invades the cell, BCV obtains some
host marker molecules through interaction with lysosome (Lys) and endosomes, matures
the endosomes in the membrane bound vacuoles, and forms acidified endosomes. At
this time, BCV is called endosomal Brucella containing vacuole (eBCV). As BCV develops
and matures, the Type IV secretory system (T4SS) mediates the interaction between the
effector protein and the endoplasmic reticulum (ER) exit site, and obtains ER and Golgi
apparatus-derived membranes. After losing the early host marker molecules, the eBCV
obtained Lys marker molecules (such as Rab7, LAMP-1, etc.) [
11
]. The BCV escaping
Lys degradation will reach the ER and fuse with the ER in a Sar1 and Rab2 dependent
manner [
12
]. At this time, the BCV is called repetitive Brucella containing vacuole (rBCV).
At the later stage of infection, rBCV will be transformed into autophagic Brucella containing
vacuole (aBCV) (Figure 1). At this time, the aBCV will not continue to mature and kill cells.
Thus far, Brucella completes the intracellular circulation, and the organism finally releases
pathogens through lysis and nonlysis mechanisms [13].
Brucella interacts with lipid rafts on the plasma membrane, which promotes Brucella to
contact with the host cells and mediates its internalization into phagocytes. The lipid rafts
contain glycosphingolipids and cholesterol, which can promote the membrane-related bio-
logical processes, such as the formation of polybasic membrane complexes, transmembrane
signaling and membrane fusion [
14
]. LPS is a key molecule in the interaction between
Brucella and host cell lipid rafts, and can prevent complement mediated bacterial lysis and
host cell apoptosis [
15
]. It has been shown that class A scavenger receptor (SR-A) and
prion protein (Pr Pc) are involved in the process of Brucella invading cells through lipid
rafts [
16
,
17
]. Prion protein and SR-A, as receptor proteins of heat shock protein 60 (HSP60)
and LPS, exist in specific lipid rafts. The destruction of lipid rafts can effectively reduce
the early survival of Brucella in macrophages, indicating that the introduction of lipid rafts
is a necessary condition for the early survival of bacteria [
18
]. Brucella enter the cell to
form phagosome and participate in the endocytosis pathway, but Brucella can be quickly
separated from the phagosome, indicating that the early survival of Brucella is related to
the lipid raft mediated signaling pathway [
19
]. In the process of Brucella’s intracellular
circulation, it needs aBCV to complete the intracellular life cycle and cell–cell diffusion [
20
].
The host protein (Yip1A) plays an important role in the formation of rBCV and aBCV. In
Yip1A knockout cells, Brucella could not form rBCV, so it could only remain in the Lys body.
The formation of aBCV depends on the small GTP enzyme Rab9 [
13
]. When the ER Beclin1
Int. J. Mol. Sci. 2021,22, 3673 3 of 13
and PI3K form a complex, rBCV begins to transform into aBCV, but with the consumption
of ATG14L, the formation of aBCV decreases gradually. By interacting with the conserved
oligomeric Golgi (COG), the effector protein BspB regulates the COG dependent transport,
reorients the golgi body derived vesicles to BCV, promotes the formation of rBCV, and
promotes the intracellular proliferation of Brucella.
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW 3 of 14
Figure 1. Brucella interacts with lipid rafts on the surface of cell membrane to enter macrophages
and form Brucella-containing vacuoles (BCV). The early BCV in macrophages is called eBCV, it ac-
quires some host marker molecules. With the maturation of eBCV, eBCV loses the marker of early
endosome, and obtains the marker molecules of late endosome and lysosome recognition, so as to
promote the fusion of eBCV and lysosome. Part of the eBCV escaped lysosome degradation and
reached the ER, and then fused with the ER by Sar1 and Rab2 to form rBCV. Brucella proliferated in
rBCV. At the late stage of infection, the rBCV containing a large number of Brucella transformed into
aBCV. The aBCV released pathogens through cleavage and noncleavage mechanisms, and the in-
tracellular circulation of Brucella ended.
Brucella interacts with lipid rafts on the plasma membrane, which promotes Brucella
to contact with the host cells and mediates its internalization into phagocytes. The lipid
rafts contain glycosphingolipids and cholesterol, which can promote the membrane-re-
lated biological processes, such as the formation of polybasic membrane complexes, trans-
membrane signaling and membrane fusion [14]. LPS is a key molecule in the interaction
between Brucella and host cell lipid rafts, and can prevent complement mediated bacterial
lysis and host cell apoptosis [15]. It has been shown that class A scavenger receptor (SR-
A) and prion protein (Pr Pc) are involved in the process of Brucella invading cells through
lipid rafts [16,17]. Prion protein and SR-A, as receptor proteins of heat shock protein 60
(HSP60) and LPS, exist in specific lipid rafts. The destruction of lipid rafts can effectively
reduce the early survival of Brucella in macrophages, indicating that the introduction of
lipid rafts is a necessary condition for the early survival of bacteria [18]. Brucella enter the
cell to form phagosome and participate in the endocytosis pathway, but Brucella can be
quickly separated from the phagosome, indicating that the early survival of Brucella is
related to the lipid raft mediated signaling pathway [19]. In the process of Brucella’s intra-
cellular circulation, it needs aBCV to complete the intracellular life cycle and cell–cell dif-
fusion [20]. The host protein (Yip1A) plays an important role in the formation of rBCV
and aBCV. In Yip1A knockout cells, Brucella could not form rBCV, so it could only remain
in the Lys body. The formation of aBCV depends on the small GTP enzyme Rab9 [13].
When the ER Beclin1 and PI3K form a complex, rBCV begins to transform into aBCV, but
with the consumption of ATG14L, the formation of aBCV decreases gradually. By inter-
acting with the conserved oligomeric Golgi (COG), the effector protein BspB regulates the
COG dependent transport, reorients the golgi body derived vesicles to BCV, promotes the
formation of rBCV, and promotes the intracellular proliferation of Brucella.
Figure 1.
Brucella interacts with lipid rafts on the surface of cell membrane to enter macrophages and
form Brucella-containing vacuoles (BCV). The early BCV in macrophages is called eBCV, it acquires
some host marker molecules. With the maturation of eBCV, eBCV loses the marker of early endosome,
and obtains the marker molecules of late endosome and lysosome recognition, so as to promote the
fusion of eBCV and lysosome. Part of the eBCV escaped lysosome degradation and reached the ER,
and then fused with the ER by Sar1 and Rab2 to form rBCV. Brucella proliferated in rBCV. At the late
stage of infection, the rBCV containing a large number of Brucella transformed into aBCV. The aBCV
released pathogens through cleavage and noncleavage mechanisms, and the intracellular circulation
of Brucella ended.
3. The Survival and Replication of Brucella in Cells
After Brucella invades the host cell, it circulates in the cell [
21
]. The rBCV stage is
tightly associated with bacterial proliferation (between 12 and 48 h post infection). At the
same time, rBCV obtains a large number of ER molecular markers, such as calmodulin,
calreticulin and ER protein Sec61. After Brucella infection, unfolded protein response (UPR)
is induced by secreting a variety of effector proteins. IRE1 and Yip1A mediate UPR to form a
complex at endoplasmic reticulum export sites (ERES) to selfphosphorylate [
22
]. Activated
IRE1 can promote the formation of ER derived vesicles. The ER derived vesicles fuse
with the Lys vesicles containing Brucella to form rBCV, which promote the proliferation of
Brucella by continuously fusing with the secretory ER derived vesicles. Brucella can survive
and propagate in the host cell mainly by the action of LPS, OMP, T4SS, two-component
regulatory system and other virulence factors [
23
,
24
]. These factors are necessary for
Brucella to invade host cells and survive and replicate in cells.
As the virulence factor of Brucella, the T4SS encoded by VirB can affect the survival
and replication of Brucella (Figure 2). VirB is a virulence gene of Brucella. Its expression in
host cells can affect the intracellular survival and replication of Brucella. The functions of
the 12 genes of VirB operon are different. At present, the effects of the 12 VirB genes on the
virulence of Brucella are mainly focused on Brucella. abortus,Brucella. melitensis and Brucella.
ovis (Table 1).
Int. J. Mol. Sci. 2021,22, 3673 4 of 13
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW 4 of 14
3. The Survival and Replication of Brucella in Cells
After Brucella invades the host cell, it circulates in the cell [21]. The rBCV stage is
tightly associated with bacterial proliferation (between 12 and 48 h post infection). At the
same time, rBCV obtains a large number of ER molecular markers, such as calmodulin,
calreticulin and ER protein Sec61. After Brucella infection, unfolded protein response (UPR)
is induced by secreting a variety of effector proteins. IRE1 and Yip1A mediate UPR to
form a complex at endoplasmic reticulum export sites (ERES) to selfphosphorylate [22].
Activated IRE1 can promote the formation of ER derived vesicles. The ER derived vesicles
fuse with the Lys vesicles containing Brucella to form rBCV, which promote the prolifera-
tion of Brucella by continuously fusing with the secretory ER derived vesicles. Brucella can
survive and propagate in the host cell mainly by the action of LPS, OMP, T4SS, two-com-
ponent regulatory system and other virulence factors [23,24]. These factors are necessary
for Brucella to invade host cells and survive and replicate in cells.
As the virulence factor of Brucella, the T4SS encoded by VirB can affect the survival
and replication of Brucella (Figure 2). VirB is a virulence gene of Brucella. Its expression in
host cells can affect the intracellular survival and replication of Brucella. The functions of
the 12 genes of VirB operon are different. At present, the effects of the 12 VirB genes on
the virulence of Brucella are mainly focused on Brucella. abortus, Brucella. melitensis and
Brucella. ovis (Table 1).
Figure 2. T4SS of Brucella is a multiprotein complex encoded by VirB operon, which participates in
the intracellular activities of Brucella. The T4SS is mainly divided into the following five parts: the
elongation region is composed of VirB2; the central and outer membrane regions were composed
of VirB7, VirB9 and VirB10; the junction region is composed of VirB5 and VirB10; the intimal re-
gion was composed of VirB3, VirB4, VirB6, VirB8 and VirB10; ATP energy region composed of
VirB4 and VirB11.
Figure 2.
T4SS of Brucella is a multiprotein complex encoded by VirB operon, which participates in
the intracellular activities of Brucella. The T4SS is mainly divided into the following five parts: the
elongation region is composed of VirB2; the central and outer membrane regions were composed
of VirB7, VirB9 and VirB10; the junction region is composed of VirB5 and VirB10; the intimal region
was composed of VirB3, VirB4, VirB6, VirB8 and VirB10; ATP energy region composed of VirB4
and VirB11.
Table 1.
The 12 genes of VirB operon and their respective function Brucella. abortus,Brucella.melitensis and Brucella.
Ovis [2529].
Operon
Whether or Not the Virulence Decreases
after Deletion Function
Brucella.
abortus
Brucella.
melitensis Brucella. ovis
VirB1 decrease unknown unknown Dissolves glycosyltransferase, which makes T4SS
easier to assemble and assemble.
VirB2 decrease decrease decrease Participate in the immune protection of the body and
affect the production of immune protection.
VirB3, VirB6,
VirB7, VirB10 decrease unknown unknown Signal transmission of bacterial transmembrane
proteins.
VirB4 decrease decrease unknown Transport substances and effectively prevent BCV and
dissolution Enzyme fusion.
VirB5, VirB8 decrease unknown unknown Regulation of intracellular transport in Brucella.
VirB9 decrease decrease unknown
As a part of the outer membrane of Brucella type IV
secretion system, it can stimulate the body to produce
immune responsean.
VirB11 decrease unknown unknown It provides energy for the secretion process of Brucella.
VirB12 unchanged unchanged unchanged Immune antigen.
Int. J. Mol. Sci. 2021,22, 3673 5 of 13
In the process of Brucella cell replication, T4SS plays an important role in a series
of processes [
30
], such as late intracellular and lysozyme markers, the recognition of
ER markers and acting on the secretion pathway, the recognition of autophagy markers,
resistance to the harsh intracellular environment and the regulation of the activation of
the immune pathway [
30
]. With the help of T4SS, Brucella transfers to ER and produces
acidic BCV under the induction of expression of VirB operon encoded by T4SS. The acidic
environment of BCV is conducive to the survival of Brucella [
31
]. BCV acidification is
an important step in the maturation of BCV, and acidic environment contributes to the
expression of VirB operon in Brucella [
32
]. Brucella induces the expression of VirB operon
in acidic environment and controls the expression of genes related to T4SS. Brucella uses
T4SS to transport effector from the membrane space to the host cell cytoplasm, so as to
regulate the signal transduction of the host cell to facilitate its survival in the host cell. In
addition to T4SS, the two-component regulatory system (BvrS/BvrR), cyclic
β
-glucan, Lux
R-like transcriptional regulator (VjbR), LPS, flagellumlike structure, and transporter-like
protein (Baca) and phosphatidylcholine (PC) are necessary components for Brucella to
invade cells and live in cells [
33
]. Cd98hc transmembrane protein also plays an important
role in intracellular proliferation and signal pathway regulation [34].
Brucella is similar to other intracellular bacteria, it infection of the host cells includes
adhesion, invasion, establishment of infection and diffusion. The interaction between host
and LPS may play an important role in Brucella’s cell viability. The essential gene (manB,
wboA) for the O-side chain synthesis of LPS is a necessary factor for Brucella to establish
the replication region in the cells [
35
]. Smooth Brucella glabrata inhibits host cell apoptosis
and promotes its survival and replication in host cells through the interaction of the o-chain
and TNF-
α
[
36
]. Thus, dead cells do not release specific factors, therefore they do not
activate the immune system and Brucella are able to avoid host immune surveillance [
37
].
In this way, Brucella enters its replication niche and proliferates in the host cells. In addition,
LPS is considered to be the main determinant of virulence, and the survival rate of the
LPS-deficient strains in the host cells is reduced. Brucella can not only escape from immune
surveillance, but also adapt to the internal environment of phagocytes and nonphagocytes
and survive in the cell. Based on the inhibition of the phagosomal–lysosomal maturation
pathway and the maladjustment of the intracellular transport, Brucella enters the cell and
then reaches the ER-derived replication niche [38].
The propagation of Brucella in cells includes two stages: the stable stage and the
exponential stage. The physiological state of the stable stage is favorable for Brucella to
adapt to the harsh living conditions in the phagocytosis body, while the exponential stage
is used to replicate under the appropriate environmental conditions, and this adaptive
regulation is completed by molecular mechanism. It has been found that the expression
of VirB operon is high in the exponential growth stage, but it is inhibited after entering
the stable stage [
39
]. Like other intracellular bacteria, Brucella have adapted to their
intracellular lifestyle and no longer need to accumulate energy storage molecules [
40
].
Brucella can survive opsonin mediated phagocytosis and replicate in cells [
41
]. The presence
of cytochromes (such as cytochrome bc1 complex or hydroquinone) with high oxygen
affinity play an important role in Brucella’s adaptation to intracellular survival. It has
been confirmed that Brucella uses heme iron polypeptide as an iron source in vitro. When
Brucella replicates in trophoblast cells, heme iron polypeptide is needed to participate.
Macrophages are the preferred host cells of Brucella, which are very important for the
heme iron polypeptide cycle in mammals [
41
]. The mutations of iron regulatory genes can
weaken the virulence of Brucella in mammalian host cells and make it more sensitive to
oxidative damage, which indicates that these mutations are involved in the growth and
intracellular survival of Brucella [42].
Int. J. Mol. Sci. 2021,22, 3673 6 of 13
4. Brucella Evades Killing of Host Immune System
Innate immunity and adaptive immunity play an important role in host immunity
against Brucella. Innate cellular immunity antiinfection effect is not good; the host immune
response caused by Brucella invasion is mainly adaptive cellular immunity [38].
4.1. Brucella Interferes with Innate Immune Recognition and Response of Host
As the first immune defense line, innate immune response plays a very important
role in the process of protecting the body from pathogens [
43
]. Brucella has been living in
cells for a long time and Brucella has evolved many mechanisms that interfere with innate
immune recognition and response in its interaction with the body (Figure 3).
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW 6 of 14
maturation pathway and the maladjustment of the intracellular transport, Brucella enters
the cell and then reaches the ER-derived replication niche [38].
The propagation of Brucella in cells includes two stages: the stable stage and the ex-
ponential stage. The physiological state of the stable stage is favorable for Brucella to adapt
to the harsh living conditions in the phagocytosis body, while the exponential stage is
used to replicate under the appropriate environmental conditions, and this adaptive reg-
ulation is completed by molecular mechanism. It has been found that the expression of
VirB operon is high in the exponential growth stage, but it is inhibited after entering the
stable stage [39]. Like other intracellular bacteria, Brucella have adapted to their intracel-
lular lifestyle and no longer need to accumulate energy storage molecules [40]. Brucella
can survive opsonin mediated phagocytosis and replicate in cells [41]. The presence of
cytochromes (such as cytochrome bc1 complex or hydroquinone) with high oxygen affin-
ity play an important role in Brucella’s adaptation to intracellular survival. It has been
confirmed that Brucella uses heme iron polypeptide as an iron source in vitro. When Bru-
cella replicates in trophoblast cells, heme iron polypeptide is needed to participate. Mac-
rophages are the preferred host cells of Brucella, which are very important for the heme
iron polypeptide cycle in mammals [41]. The mutations of iron regulatory genes can
weaken the virulence of Brucella in mammalian host cells and make it more sensitive to
oxidative damage, which indicates that these mutations are involved in the growth and
intracellular survival of Brucella [42].
4. Brucella Evades Killing of Host Immune System
Innate immunity and adaptive immunity play an important role in host immunity
against Brucella. Innate cellular immunity antiinfection effect is not good; the host immune
response caused by Brucella invasion is mainly adaptive cellular immunity [38].
4.1. Brucella Interferes with Innate Immune Recognition and Response of Host
As the first immune defense line, innate immune response plays a very important
role in the process of protecting the body from pathogens [43]. Brucella has been living in
cells for a long time and Brucella has evolved many mechanisms that interfere with innate
immune recognition and response in its interaction with the body (Figure 3).
Figure 3. Brucella can inhibit the secretion of IL-2 by antigen presenting cells, and then prevent natural killer cells (NK)
from secreting inflammatory factors such as IFN-γ and TNF-α. Brucella can also inhibit IFN-γ mediated phagocytosis to
Figure 3.
Brucella can inhibit the secretion of IL-2 by antigen presenting cells, and then prevent natural killer cells (NK) from
secreting inflammatory factors such as IFN-γand TNF-α.Brucella can also inhibit IFN-γmediated phagocytosis to escape
the killing of immune system. Brucella affects the maturation of DC by blocking TLR2 receptor pathway, and interferes with
the establishment of Th1 immune response by inhibiting macrophages to reduce IL-12 secretion and preventing DC from
activating T-lymphocytes.
The innate immune system recognizes microbes by characteristic molecules like the
Gram-negative LPS Lipid A (the LPS bioactive moiety) signals through Toll-like receptors
(TLRs) to induce proinflammatory molecules and small GTPases of the p47 family involved
in intracellular pathogen control. Brucella LPS exhibits a low toxicity and its atypical
structure was postulated to delay the host immune response, favouring the establishment
of chronic disease. Brucella Lipid A is a 2,3-diaminoglucose disaccharide substituted with
C16, C18, C28 and other very long acyl chains. This peculiar structure is a poor agonist of
TLR4/myeloid differentiation-2 (MD-2) and therefore a paradigm has emerged proposing
Brucella LPS as a crucial virulence factor that hampers recognition by pattern recognition
receptors (PRR) and plays essential roles during infection. Research shows that Brucella
LPS did not induce inflammatory responses in macrophages and DCs, two of the most
important sentinels of the immune system.This was attributed to its poor recognition by
TLR4/MD-2, which is widely considered to be the major receptor complex for LPS binding
and signalling [44].
Host cells recognize the harmful substances by the interaction of PRR and pathogen
associated molecular pattern (PAMPs), so as to stimulate the body to produce related
immune response to kill and eliminate pathogens. However, there is increasing evidence
that Brucella display altered PAMPs in key molecules, suggesting that to escape detection by
innate immunity is a survival strategy. One of the best examples of a structure with altered
PAMPs is the LPS of Brucella.Brucella LPS bears a noncanonical lipid A and, although it
Int. J. Mol. Sci. 2021,22, 3673 7 of 13
signals TLR4, it is active only at very high concentrations [
45
]. The flagellin of Brucella lacks
the specific domain recognized by TLR5 and plays an important role in immune escape [
46
].
Moreover, Brucella LPS confers a highly resistant phenotype to cationic bactericidal peptides
and makes Brucella a poor activator of the complement system [
47
]. The Lipid A of Brucella
contains a longer fatty acid chain (C28), which greatly reduces its endotoxin properties [
23
].
There are free hydroxyl residues in the O-chain of bacterial LPS, which is favorable for
binding with C3. However, the O-chain of Brucella lacks free hydroxyl. Complement C3 can
inhibit the production of C3a and C5a by contacting with the specific O-chain of Brucella
LPS, thus avoiding the capture of the host immune system. At the same time, LPS on the
surface of the cell wall has many long side chains, which prevent the membrane attacking
complex from contacting the cell membrane (Figure 4).
Int. J. Mol. Sci. 2021, 22, x FOR PEER REVIEW 7 of 14
escape the killing of immune system. Brucella affects the maturation of DC by blocking TLR2 receptor pathway, and inter-
feres with the establishment of Th1 immune response by inhibiting macrophages to reduce IL-12 secretion and preventing
DC from activating T-lymphocytes.
The innate immune system recognizes microbes by characteristic molecules like the
Gram-negative LPS Lipid A (the LPS bioactive moiety) signals through Toll-like receptors
(TLRs) to induce proinflammatory molecules and small GTPases of the p47 family in-
volved in intracellular pathogen control. Brucella LPS exhibits a low toxicity and its atyp-
ical structure was postulated to delay the host immune response, favouring the establish-
ment of chronic disease. Brucella Lipid A is a 2,3-diaminoglucose disaccharide substituted
with C16, C18, C28 and other very long acyl chains. This peculiar structure is a poor ago-
nist of TLR4/myeloid differentiation-2 (MD-2) and therefore a paradigm has emerged pro-
posing Brucella LPS as a crucial virulence factor that hampers recognition by pattern
recognition receptors (PRR) and plays essential roles during infection. Research shows
that Brucella LPS did not induce inflammatory responses in macrophages and DCs, two
of the most important sentinels of the immune system.This was attributed to its poor
recognition by TLR4/MD-2, which is widely considered to be the major receptor complex
for LPS binding and signalling [44].
Host cells recognize the harmful substances by the interaction of PRR and pathogen
associated molecular pattern (PAMPs), so as to stimulate the body to produce related im-
mune response to kill and eliminate pathogens. However, there is increasing evidence
that Brucella display altered PAMPs in key molecules, suggesting that to escape detection
by innate immunity is a survival strategy. One of the best examples of a structure with
altered PAMPs is the LPS of Brucella. Brucella LPS bears a noncanonical lipid A and, alt-
hough it signals TLR4, it is active only at very high concentrations [45]. The flagellin of
Brucella lacks the specific domain recognized by TLR5 and plays an important role in im-
mune escape [46]. Moreover, Brucella LPS confers a highly resistant phenotype to cationic
bactericidal peptides and makes Brucella a poor activator of the complement system [47].
The Lipid A of Brucella contains a longer fatty acid chain (C28), which greatly reduces its
endotoxin properties [23]. There are free hydroxyl residues in the O-chain of bacterial LPS,
which is favorable for binding with C3. However, the O-chain of Brucella lacks free hy-
droxyl. Complement C3 can inhibit the production of C3a and C5a by contacting with the
specific O-chain of Brucella LPS, thus avoiding the capture of the host immune system. At
the same time, LPS on the surface of the cell wall has many long side chains, which prevent
the membrane attacking complex from contacting the cell membrane (Figure 4).
Figure 4. As an important virulence factor of Brucella, LPS plays an important role in escaping from the host immune
system. The acetyl side chain (C28) of Brucella LPS reduces the nature of endotoxin to avoid the recognition of Toll-like
receptor 4 (TLR4), thus avoiding the monitoring of host immune system. The specific O-chain contained in Brucella LPS
can inhibit the production of C3a and C5a by complement C3, and then inhibit the degranulation of neutrophils, so as to
Figure 4.
As an important virulence factor of Brucella, LPS plays an important role in escaping from the host immune
system. The acetyl side chain (C28) of Brucella LPS reduces the nature of endotoxin to avoid the recognition of Toll-like
receptor 4 (TLR4), thus avoiding the monitoring of host immune system. The specific O-chain contained in Brucella LPS
can inhibit the production of C3a and C5a by complement C3, and then inhibit the degranulation of neutrophils, so as to
prevent the release of myeloperoxidase (MPO) and other lysosomal substances, and prevent them from being captured by
the host immune system.
It was found that the removal of polymorphonuclear before the start of adaptive
immunity was helpful to the elimination of bacteria in mice, indicating that neutrophils
inhibited the immune response of the body to Brucella [
48
,
49
]. Brucella also has different re-
sistance mechanisms to phospholipase A2, catheteridin, Lys and defensins, so as to ensure
its transport in lymphoid tissue. Brucella induces antigen-presenting cells to secrete IL-2
and activate NK cells. NK cells secrete IFN-
γ
, TNF-
α
, GM-CSF and other cytokines, which
play an important role in Th1 and Tc1 reactions [
50
]. Brucella evades the host immune
response by affecting macrophage function. Brucella can inhibit IFN-
γ
mediated phago-
cytosis and TNF-
α
expression in macrophages [
51
]. Infected macrophages can produce
proinflammatory factors (TNF-
α
, IL-6, IL-12) and inflammatory chemokines (GRO-
α
, IL-8,
MCP-1). TNF-
α
can significantly enhance the bactericidal ability of macrophages, IL-12
can induce Th1 immune response and produce IFN-
γ
.Brucella regulates the expression of
MHC-I and MHC-II by regulating IFN-
γ
secretion. IFN-
γ
mediated Th1 immune response
is essential for Brucella clearance [
52
]. Brucella can affect the maturation of DC by blocking
the TLR2 receptor pathway, and interfere with the establishment of type Th1 immune
response by reducing the secretion of IL-12 and preventing the activation of T-lymphocyte
by DC [53].
4.2. Transmission Mechanism of Brucella Interfering Host Adaptive Immune Response
The adaptive immune response of the organism mainly includes the humoral immune
response and the cellular immune response [
54
]. In the long-term evolution, Brucella has
Int. J. Mol. Sci. 2021,22, 3673 8 of 13
produced the transmission mechanism of interfering information from innate immunity to
acquired immunity, so as to establish chronic infection [49].
The decrease in macrophage and DCs recruitment after Brucella infection leads to the
decrease of CD8+ T lymphocyte activation, which leads to the formation of immunosup-
pression, which is conducive to Brucella replication and chronic infection [
55
]. Brucella
lumazine synthase (BLS) transmits signals through TLR4 and induces DC maturation
and CD8+ T-lymphocytel toxicity, so as to inhibit tumor growth and regulate innate and
adaptive immune responses. As one of several virulence factors necessary for Brucella to
establish chronic infection, prpA can interact with macrophages to promote B-cell pro-
liferation [
56
]. PrpA can regulate IFN-
γ
, TNF-
α
, IL-10 and TGF-
β
1 in the early stage of
infection [
57
]. The results showed that prpA, Btp1/TcpB and LPS, as immunomodulators,
had the ability to inhibit IFN-
γ
secretion and promote IL-10 secretion, thus affecting Th1
immune response [58].
Brucella effectors can control the TLR signaling pathway involved in DC maturation,
and have a significant effect on T lymphocyte activation and antigen presentation. The
sequence of Brucella TIR protein 1 (btp1) is similar to toll/IL-1 domain family, because
of the significance of the TIR domain in TLR signal [
23
,
59
]. BTP 1 not only inhibited
the production of proinflammatory cytokines, but also inhibited the maturation of DC,
resulting in the inhibition of TLR2 and TLR4 signals [60].
Brucella inhibits immune signal transduction by expressing secreted proteins contain-
ing the TIR domain Btp1/TcpB [
61
]. The detailed mechanism of this protein is still not fully
understood, but there is evidence that when it binds to the TIR domain-containing adaptor
protein (TIRAP/Mal), it competes with myeloid differentiation response gene 88 (MyD88),
which not only promotes TIRAP/Mal ubiquitination degradation but also inhibits TLR4
and TLR2 signal transduction [
62
]. In this way, Brucella inhibits DC maturation and the
production of proinflammatory cytokines IL-12 and TNF-
α
. In addition, this protein also
inhibits the killing effect of CD8+ T-lymphocyte on Brucella target cells [63].
5. Chronic Infection Caused by Brucella
After Brucella invade the macrophage, with the proliferation of BCV in ER, some
components of Brucella can interact with host cells, which can inhibit the synthesis of
bactericides, affect the activation of signal transduction pathway, induce super allergic
reaction, and finally cause systemic persistent infection [
64
]. When the T4SS encoded by
VirB stimulates the body, the immune response produced by the host can inhibit the growth
and reproduction of Brucella, but it cannot kill Brucella, resulting in a confrontation between
Brucella and the host, causing the body to develop persistent infection [65].
The interaction between Brucella and host immune system is very important in caus-
ing persistent infection, but immune evasion is not the only mechanism. Studies have
shown that the genes needed for Brucella persistence are related to changes in bacterial
metabolism and the ability of pathogens to utilize specific nutrients [
66
]. The macrophage
is the main target cell of Brucella persistence, which provides a place for Brucella replication
and survival [
67
]. The key to understanding Brucella’s cell viability and chronic infection
is to understand the interaction between Brucella and different macrophage subsets [
68
].
Several factors required for the long-term existence of Brucella in the host cannot medi-
ate the replication of Brucella in
in vitro
cultured macrophages, indicating that different
macrophage populations and their metabolic status may be the determinants of chronic
diseases [66,69].
Brucella not only has the ability to resist phagocyte sterilization, but also can prevent
antigen-specific T-lymphocyte from recognizing themselves, thus forming a microenvi-
ronment conducive to their survival and reproduction, leading to chronic persistent infec-
tion [
28
]. The proliferation and replication of Brucella weakens the phagocytic function of
macrophages, which results in the loss of the cell killing and antigen presenting functions of
macrophages, thus avoiding the host’s immune defense mechanism and causing persistent
infection [
70
]. Persistent infection is determined not only by Brucella’s ability to evade host
Int. J. Mol. Sci. 2021,22, 3673 9 of 13
immune response, but also by its ability to utilize available nutrients in the chronic phase
of infection. Macrophages play an important role in host physiology and metabolism [
71
],
which are the key to the survival of Brucella.Brucella not only has the ability to resist the
phagocytic bactericidal effect, but also makes the host cells form a microenvironment which
is conducive to its survival and reproduction. This microenvironment can make Brucella
live in the host cell for a long time, and eventually lead to chronic persistent infection.
The macrophage is the main target cell of Brucella.Brucella can inhibit the apoptosis
of the macrophage by inhibiting the secretion of TNF-
α
so that it can survive in the cell
and multiply in large numbers, weaken the phagocytic function of macrophage, lose the
killing effect and antigenic presenting function of macrophage, and finally escape the
immune surveillance of host [
72
]. On the other hand, macrophages have a weak ability to
activate the initial T-lymphocyte and do not express CD1 molecules with antigen presenting
function, so they cannot effectively present lipid antigens to NK cells, which may be the
reason for the formation of Brucella persistent infection. Brucella regulates the immune
response by inducing regulatory cytokines (such as IL-10), which indicates that the IL-10
pathway plays an important role in the chronic infection caused by Brucella [7375].
Brucella evades the host’s initial natural immunity through Toll-like receptor (TLR),
and causes a slight inflammatory reaction by modifying virulent factors such as LPS and
flagellin [
51
], which causes Brucella to develop persistent infection in the body [
23
]. In
addition, Brucella can regulate immune response by inducing regulatory cytokines (such
as IL-10), indicates that IL-10 pathway can play an important role in bacterial persistent
infection [
73
75
]. It is worth noting that Brucella IL-10 inhibits the bactericidal ability
of macrophages activated by IFN-
γ
and the production of proinflammatory cytokine
in vitro [74,75].
Brucella can induce CD4(+) CD25(+) T-lymphocyte to produce anti-inflammatory cy-
tokine IL-10, thus inhibiting the immune activation of macrophages. The early production
of IL-10 by CD25 (+) CD4 (+) T-lymphocyte can regulate the function of macrophages
and help to promote the initial balance between proinflammatory cytokines and anti-
inflammatory cytokines beneficial to pathogens, so as to promote the survival and persis-
tent infection of bacteria [
76
]. Proline racemase of Brucella acts as mitogen of B lymphocytes
and induces spleen cells to secrete IL-10, which may be the basis of its persistent infec-
tion of mononuclear phagocyte system in mice [
77
]. In conclusion, these data indicate
that IL-10 plays an important role in regulating the initial immune response. Brucella
infection can increase the survival and persistent infection of pathogens by regulating
macrophage function.
Trophoblast cells are also the key target cells for the survival and replication of Brucella.
The replication ability of Brucella in host cells is the core of its pathogenicity. It was
found that the accumulation of Brucella in trophoblast cells was higher than that in other
organs [
78
]. In the extravillous trophoblast (EVTs), Brucella abortus forms an acidified
lysosomal vesicle in which it proliferates and replicates. These vesicles play an important
role in the chronic infection caused by Brucella [
79
]. It was found that the invasion of
Brucella to trophoblast cells mainly caused cell necrosis and released a large number of
Brucella. The necrotic foci of Brucella spread continuously through the capillaries and
became the main source of chronic infection of the placenta [80].
It has been found that the proliferation of Brucella in trophoblast cells is related to
the ER network, and Brucella establishes a proliferative living environment through the
interaction with the endoplasmic network [
81
]. Brucella abortus invades and replicates in the
human trophoblastic cell line Swan-71 and that the intracellular survival of the bacterium
depends on a functional VirB operon. After Brucella infection, the trophoblast cells of
ruminants can produce a lot of erythritol, which can promote the growth and reproduction
of bacteria. At present, little is known about the changes of the trophoblast environment
after Brucella infection, which need further research and discovery.
Int. J. Mol. Sci. 2021,22, 3673 10 of 13
6. Conclusions
The relationship between intracellular bacteria and host is very complex, involving a
variety of biological factors and signal pathways. As a facultative intracellular bacteria,
Brucella can colonize, grow and reproduce for a long time after invading the host cell, which
is completely dependent on its survival cycle in the host cell and the function of escaping
from the host immune system. Although the research on the facultative intracellular
parasitism mechanism of Brucella is still in progress, there are still many problems to be
further explained.
Author Contributions:
Conceptualization, H.J. and Z.Z.; methodology, Y.X.; software, B.L.; valida-
tion, H.J., Z.Z. and B.L.; formal analysis, M.L.; investigation, H.Z.; resources, X.G.; data curation,
G.G.; writing—original draft preparation, Z.Z.; writing—review and editing, H.J.; visualization, B.L.;
supervision, Y.X.; project administration, H.J.; funding acquisition, H.J. All authors have read and
agreed to the published version of the manuscript.
Funding:
This work was financially supported by the National Science Foundation for Young Scien-
tists of China (No. 31802215), the Natural Science Foundation of Chongqing (No. cstc2018jcyjA0807,
cstc2020jcyj-msxm0522), and the Fundamental Research Funds for the Central Universities (Nos.
XDJK2020C022, XDJK2019C024).
Acknowledgments:
We are very grateful to Jianjun Wen from University of Texas Medical Branch
for helpful discussions during the preparation of this manuscript.
Conflicts of Interest: The authors declare no conflict of interest.
References
1.
Pappas, G.; Papadimitriou, P.; Akritidis, N.; Christou, L.; Tsianos, E.V. The new global map of human Brucellosis. Lancet Infect.
Dis. 2006,6, 91–99. [CrossRef]
2.
Pekpak, E.; Sirvan Cetin, B. Secondary Hemophagocytic Lymphohistocytosis in a Child with Brucellosis. J. Pediatr. Hematol.
Oncol. 2017,39, e501–e503. [CrossRef]
3. Delrue, R.M.; Martinez-Lorenzo, M.; Lestrate, P.; Danese, I.; Bielarz, V.; Mertens, P.; De Bolle, X.; Tibor, A.; Gorvel, J.P.; Letesson,
J.J. Identification of Brucella spp. genes involved in intracellular trafficking. Cell Microbiol. 2001,3, 487–497. [CrossRef]
4.
Al Dahouk, S.; Sprague, L.D.; Neubauer, H. New developments in the diagnostic procedures for zoonotic Brucellosis in humans.
Rev. Sci. Tech. 2013,32, 177–188. [CrossRef]
5.
Mantur, B.G.; Amarnath, S.K.; Shinde, R.S. Review of clinical and laboratory features of human Brucellosis. Indian J. Med.
Microbiol. 2007,25, 188–202. [CrossRef] [PubMed]
6.
Diaz Aparicio, E. Epidemiology of Brucellosis in domestic animals caused by Brucella melitensis,Brucella suis and Brucella abortus.
Rev. Sci Tech. 2013,32, 43–51; discussion 53–60. [CrossRef] [PubMed]
7. Cardoso, P.G.; Macedo, G.C.; Azevedo, V.; Oliveira, S.C. Brucella spp noncanonical LPS: Structure, biosynthesis, and interaction
with host immune system. Microb. Cell Fact. 2006,5, 13. [CrossRef] [PubMed]
8.
Weynants, V.; Tibor, A.; Denoel, P.A.; Saegerman, C.; Godfroid, J.; Thiange, P.; Letesson, J.J. Infection of cattle with Yersinia
enterocolitica O:9 a cause of the false positive serological reactions in bovine Brucellosis diagnostic tests. Vet. Microbiol.
1996
,48,
101–112. [CrossRef]
9.
Copin, R.; Vitry, M.A.; Hanot Mambres, D.; Machelart, A.; De Trez, C.; Vanderwinden, J.M.; Magez, S.; Akira, S.; Ryffel, B.; Carlier,
Y.; et al. In situ microscopy analysis reveals local innate immune response developed around Brucella infected cells in resistant
and susceptible mice. PLoS Pathog. 2012,8, e1002575. [CrossRef] [PubMed]
10.
Kohler, S.; Michaux-Charachon, S.; Porte, F.; Ramuz, M.; Liautard, J.P. What is the nature of the replicative niche of a stealthy bug
named Brucella?Trends Microbiol. 2003,11, 215–219. [CrossRef]
11.
von Bargen, K.; Gorvel, J.P.; Salcedo, S.P. Internal affairs: Investigating the Brucella intracellular lifestyle. FEMS Microbiol. Rev.
2012,36, 533–562. [CrossRef] [PubMed]
12.
Celli, J.; de Chastellier, C.; Franchini, D.M.; Pizarro-Cerda, J.; Moreno, E.; Gorvel, J.P. Brucella evades macrophage killing via
VirB-dependent sustained interactions with the endoplasmic reticulum. J. Exp. Med. 2003,198, 545–556. [CrossRef]
13.
Starr, T.; Child, R.; Wehrly, T.D.; Hansen, B.; Hwang, S.; Lopez-Otin, C.; Virgin, H.W.; Celli, J. Selective subversion of autophagy
complexes facilitates completion of the Brucella intracellular cycle. Cell Host Microbe 2012,11, 33–45. [CrossRef] [PubMed]
14.
Cutler, S.J.; Whatmore, A.M.; Commander, N.J. Brucellosis–new aspects of an old disease. J. Appl. Microbiol.
2005
,98, 1270–1281.
[CrossRef] [PubMed]
15.
Jimenez de Bagues, M.P.; Terraza, A.; Gross, A.; Dornand, J. Different responses of macrophages to smooth and rough Brucella
spp.: Relationship to virulence. Infect. Immun. 2004,72, 2429–2433. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2021,22, 3673 11 of 13
16.
Kim, S.; Watarai, M.; Suzuki, H.; Makino, S.; Kodama, T.; Shirahata, T. Lipid raft microdomains mediate class A scavenger
receptor-dependent infection of Brucella abortus.Microb. Pathog. 2004,37, 11–19. [CrossRef]
17.
Watarai, M.; Kim, S.; Erdenebaatar, J.; Makino, S.; Horiuchi, M.; Shirahata, T.; Sakaguchi, S.; Katamine, S. Cellular prion protein
promotes Brucella infection into macrophages. J. Exp. Med. 2003,198, 5–17. [CrossRef]
18.
Naroeni, A.; Porte, F. Role of cholesterol and the ganglioside GM(1) in entry and short-term survival of Brucella suis in murine
macrophages. Infect. Immun. 2002,70, 1640–1644. [CrossRef]
19.
Porte, F.; Naroeni, A.; Ouahrani-Bettache, S.; Liautard, J.P. Role of the Brucella suis lipopolysaccharide O antigen in phagosomal
genesis and in inhibition of phagosome-lysosome fusion in murine macrophages. Infect. Immun.
2003
,71, 1481–1490. [CrossRef]
20.
Starr, T.; Ng, T.W.; Wehrly, T.D.; Knodler, L.A.; Celli, J. Brucella intracellular replication requires trafficking through the late
endosomal/lysosomal compartment. Traffic 2008,9, 678–694. [CrossRef]
21. Celli, J. The Intracellular Life Cycle of Brucella spp. Microbiol. Spectr. 2019,7. [CrossRef]
22.
Gorvel, J.P.; Moreno, E. Brucella intracellular life: From invasion to intracellular replication. Vet. Microbiol.
2002
,90, 281–297.
[CrossRef]
23.
Atluri, V.L.; Xavier, M.N.; de Jong, M.F.; den Hartigh, A.B.; Tsolis, R.M. Interactions of the human pathogenic Brucella species
with their hosts. Annu. Rev. Microbiol. 2011,65, 523–541. [CrossRef] [PubMed]
24. Byndloss, M.X.; Tsolis, R.M. Brucella spp. Virulence Factors and Immunity. Annu Rev. Anim Biosci 2016,4, 111–127. [CrossRef]
25.
Hoppner, C.; Carle, A.; Sivanesan, D.; Hoeppner, S.; Baron, C. The putative lytic transglycosylase VirB1 from Brucella suis interacts
with the type IV secretion system core components VirB8, VirB9 and VirB11. Microbiology (Reading)
2005
,151 Pt 11, 3469–3482.
[CrossRef]
26.
den Hartigh, A.B.; Sun, Y.H.; Sondervan, D.; Heuvelmans, N.; Reinders, M.O.; Ficht, T.A.; Tsolis, R.M. Differential requirements
for VirB1 and VirB2 during Brucella abortus infection. Infect. Immun. 2004,72, 5143–5149. [CrossRef] [PubMed]
27.
Rouot, B.; Alvarez-Martinez, M.T.; Marius, C.; Menanteau, P.; Guilloteau, L.; Boigegrain, R.A.; Zumbihl, R.; O’Callaghan, D.;
Domke, N.; Baron, C. Production of the type IV secretion system differs among Brucella species as revealed with VirB5- and
VirB8-specific antisera. Infect. Immun. 2003,71, 1075–1082. [CrossRef] [PubMed]
28.
Hare, S.; Bayliss, R.; Baron, C.; Waksman, G. A large domain swap in the VirB11 ATPase of Brucella suis leaves the hexameric
assembly intact. J. Mol. Biol. 2006,360, 56–66. [CrossRef] [PubMed]
29.
Mirkalantari, S.; Amirmozafari, N.; Kazemi, B.; Irajian, G. Molecular cloning of virB12 gene of Brucella melitensis 16M strain in
pET28a vector. Asian Pac. J. Trop. Med. 2012,5, 511–513. [CrossRef]
30.
Ke, Y.; Wang, Y.; Li, W.; Chen, Z. Type IV secretion system of Brucella spp. and its effectors. Front. Cell Infect. Microbiol.
2015
,5, 72.
[CrossRef]
31.
Hanna, N.; Jimenez de Bagues, M.P.; Ouahrani-Bettache, S.; El Yakhlifi, Z.; Kohler, S.; Occhialini, A. The virB operon is essential
for lethality of Brucella microti in the Balb/c murine model of infection. J. Infect. Dis 2011,203, 1129–1135. [CrossRef] [PubMed]
32.
Porte, F.; Liautard, J.P.; Kohler, S. Early acidification of phagosomes containing Brucella suis is essential for intracellular survival
in murine macrophages. Infect. Immun. 1999,67, 4041–4047. [CrossRef]
33.
Roop, R.M., 2nd; Gaines, J.M.; Anderson, E.S.; Caswell, C.C.; Martin, D.W. Survival of the fittest: How Brucella strains adapt to
their intracellular niche in the host. Med. Microbiol. Immunol. 2009,198, 221–238. [CrossRef]
34.
Keriel, A.; Botella, E.; Estrach, S.; Bragagnolo, G.; Vergunst, A.C.; Feral, C.C.; O’Callaghan, D. Brucella Intracellular Life Relies on
the Transmembrane Protein CD98 Heavy Chain. J. Infect. Dis. 2015,211, 1769–1778. [CrossRef] [PubMed]
35.
Zygmunt, M.S.; Blasco, J.M.; Letesson, J.J.; Cloeckaert, A.; Moriyon, I. DNA polymorphism analysis of Brucella lipopolysaccharide
genes reveals marked differences in O-polysaccharide biosynthetic genes between smooth and rough Brucella species and novel
species-specific markers. BMC Microbiol. 2009,9, 92. [CrossRef] [PubMed]
36.
Glowacka, P.; Zakowska, D.; Naylor, K.; Niemcewicz, M.; Bielawska-Drozd, A. Brucella—Virulence Factors, Pathogenesis and
Treatment. Pol. J. Microbiol. 2018,67, 151–161. [CrossRef] [PubMed]
37.
Zhao, Y.; Hanniffy, S.; Arce-Gorvel, V.; Conde-Alvarez, R.; Oh, S.; Moriyon, I.; Memet, S.; Gorvel, J.P. Immunomodulatory
properties of Brucella melitensis lipopolysaccharide determinants on mouse dendritic cells
in vitro
and
in vivo
.Virulence
2018
,9,
465–479. [CrossRef]
38.
Contreras-Rodriguez, A.; Ramirez-Zavala, B.; Contreras, A.; Schurig, G.G.; Sriranganathan, N.; Lopez-Merino, A. Purification
and characterization of an immunogenic aminopeptidase of Brucella melitensis.Infect. Immun. 2003,71, 5238–5244. [CrossRef]
39.
Roop, R.M., 2nd; Gee, J.M.; Robertson, G.T.; Richardson, J.M.; Ng, W.L.; Winkler, M.E. Brucella stationary-phase gene expression
and virulence. Annu Rev. Microbiol. 2003,57, 57–76. [CrossRef]
40.
Chain, P.S.; Comerci, D.J.; Tolmasky, M.E.; Larimer, F.W.; Malfatti, S.A.; Vergez, L.M.; Aguero, F.; Land, M.L.; Ugalde, R.A.; Garcia,
E. Whole-genome analyses of speciation events in pathogenic Brucellae.Infect. Immun. 2005,73, 8353–8361. [CrossRef]
41.
Bellaire, B.H.; Elzer, P.H.; Baldwin, C.L.; Roop, R.M. Production of the siderophore 2,3-dihydroxybenzoic acid is required for
wild-type growth of Brucella abortus in the presence of erythritol under low-iron conditions
in vitro
.Infect. Immun.
2003
,71,
2927–2932. [CrossRef] [PubMed]
42.
Shim, S.; Im, Y.B.; Jung, M.; Park, W.B.; Yoo, H.S. Genes Related to Intracellular Survival of Brucella abortus in THP-1 Macrophage
Cells. J. Microbiol. Biotechnol. 2018,28, 1736–1748. [CrossRef] [PubMed]
43.
Oliveira, S.C.; de Oliveira, F.S.; Macedo, G.C.; de Almeida, L.A.; Carvalho, N.B. The role of innate immune receptors in the control
of Brucella abortus infection: Toll-like receptors and beyond. Microbes Infect. 2008,10, 1005–1009. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2021,22, 3673 12 of 13
44.
Park, B.S.; Song, D.H.; Kim, H.M.; Choi, B.S.; Lee, H.; Lee, J.O. The structural basis of lipopolysaccharide recognition by the
TLR4-MD-2 complex. Nature 2009,458, 1191–1195. [CrossRef]
45.
Lapaque, N.; Takeuchi, O.; Corrales, F.; Akira, S.; Moriyon, I.; Howard, J.C.; Gorvel, J.P. Differential inductions of TNF-alpha
and IGTP, IIGP by structurally diverse classic and non-classic lipopolysaccharides. Cell Microbiol.
2006
,8, 401–413. [CrossRef]
[PubMed]
46.
Andersen-Nissen, E.; Smith, K.D.; Strobe, K.L.; Barrett, S.L.; Cookson, B.T.; Logan, S.M.; Aderem, A. Evasion of Toll-like receptor
5 by flagellated bacteria. Proc. Natl. Acad. Sci. USA 2005,102, 9247–9252. [CrossRef] [PubMed]
47.
Freer, E.; Moreno, E.; Moriyon, I.; Pizarro-Cerda, J.; Weintraub, A.; Gorvel, J.P. Brucella-Salmonella lipopolysaccharide chimeras
are less permeable to hydrophobic probes and more sensitive to cationic peptides and EDTA than are their native Brucella spp.
counterparts. J. Bacteriol. 1996,178, 5867–5876. [CrossRef]
48.
Barquero-Calvo, E.; Mora-Cartin, R.; Arce-Gorvel, V.; de Diego, J.L.; Chacon-Diaz, C.; Chaves-Olarte, E.; Guzman-Verri, C.; Buret,
A.G.; Gorvel, J.P.; Moreno, E. Brucella abortus Induces the Premature Death of Human Neutrophils through the Action of Its
Lipopolysaccharide. PLoS Pathog. 2015,11, e1004853. [CrossRef]
49.
Mora-Cartin, R.; Gutierrez-Jimenez, C.; Alfaro-Alarcon, A.; Chaves-Olarte, E.; Chacon-Diaz, C.; Barquero-Calvo, E.; Moreno, E.
Neutrophils Dampen Adaptive Immunity in Brucellosis. Infect. Immun. 2019,87. [CrossRef]
50.
Gao, N.; Jennings, P.; Guo, Y.; Yuan, D. Regulatory role of natural killer (NK) cells on antibody responses to Brucella abortus.Innate
Immun. 2011,17, 152–163. [CrossRef]
51.
Barrionuevo, P.; Delpino, M.V.; Velasquez, L.N.; Garcia Samartino, C.; Coria, L.M.; Ibanez, A.E.; Rodriguez, M.E.; Cassataro,
J.; Giambartolomei, G.H. Brucella abortus inhibits IFN-gamma-induced FcgammaRI expression and FcgammaRI-restricted
phagocytosis via toll-like receptor 2 on human monocytes/macrophages. Microbes Infect.
2011
,13, 239–250. [CrossRef] [PubMed]
52.
Velasquez, L.N.; Milillo, M.A.; Delpino, M.V.; Trotta, A.; Mercogliano, M.F.; Pozner, R.G.; Schillaci, R.; Elizalde, P.V.; Giambar-
tolomei, G.H.; Barrionuevo, P. Inhibition of MHC-I by Brucella abortus is an early event during infection and involves EGFR
pathway. Immunol. Cell Biol. 2017,95, 388–398. [CrossRef] [PubMed]
53.
Avila-Calderon, E.D.; Flores-Romo, L.; Sharon, W.; Donis-Maturano, L.; Becerril-Garcia, M.A.; Arreola, M.G.A.; Reynoso, B.A.;
Guemes, F.S.; Contreras-Rodriguez, A. Dendritic cells and Brucella spp. interaction: The sentinel host and the stealthy pathogen.
Folia Microbiol. (Praha) 2020,65, 1–16. [CrossRef] [PubMed]
54.
Cannella, A.P.; Tsolis, R.M.; Liang, L.; Felgner, P.L.; Saito, M.; Sette, A.; Gotuzzo, E.; Vinetz, J.M. Antigen-specific acquired
immunity in human Brucellosis: Implications for diagnosis, prognosis, and vaccine development. Front. Cell Infect. Microbiol.
2012,2, 1. [CrossRef] [PubMed]
55.
Pasquali, P.; Thornton, A.M.; Vendetti, S.; Pistoia, C.; Petrucci, P.; Tarantino, M.; Pesciaroli, M.; Ruggeri, F.; Battistoni, A.; Shevach,
E.M. CD4+CD25+ T regulatory cells limit effector T cells and favor the progression of Brucellosis in BALB/c mice. Microbes Infect.
2010,12, 3–10. [CrossRef] [PubMed]
56.
Spera, J.M.; Guaimas, F.; Corvi, M.M.; Ugalde, J.E. Brucella Hijacks Host-Mediated Palmitoylation To Stabilize and Localize PrpA
to the Plasma Membrane. Infect. Immun. 2018,86. [CrossRef]
57.
Spera, J.M.; Comerci, D.J.; Ugalde, J.E. Brucella alters the immune response in a prpA-dependent manner. Microb. Pathog.
2014
,
67–68, 8–13. [CrossRef]
58.
Kianmehr, Z.; Soleimanjahi, H.; Ardestani, S.K.; Fotouhi, F.; Abdoli, A. Influence of Brucella abortus lipopolysaccharide as an
adjuvant on the immunogenicity of HPV-16 L1VLP vaccine in mice. Med. Microbiol. Immunol. 2015,204, 205–213. [CrossRef]
59.
Salcedo, S.P.; Marchesini, M.I.; Lelouard, H.; Fugier, E.; Jolly, G.; Balor, S.; Muller, A.; Lapaque, N.; Demaria, O.; Alexopoulou,
L.; et al. Brucella control of dendritic cell maturation is dependent on the TIR-containing protein Btp1. PLoS Pathog
2008
,4, e21.
[CrossRef]
60.
Li, W.; Ke, Y.; Wang, Y.; Yang, M.; Gao, J.; Zhan, S.; Xinying, D.; Huang, L.; Li, W.; Chen, Z.; et al. Brucella TIR-like protein
TcpB/Btp1 specifically targets the host adaptor protein MAL/TIRAP to promote infection. Biochem. Biophys. Res. Commun.
2016
,
477, 509–514. [CrossRef]
61.
Kaplan-Turkoz, B.; Koelblen, T.; Felix, C.; Candusso, M.P.; O’Callaghan, D.; Vergunst, A.C.; Terradot, L. Structure of the
Toll/interleukin 1 receptor (TIR) domain of the immunosuppressive Brucella effector BtpA/Btp1/TcpB. FEBS Lett.
2013
,587,
3412–3416. [CrossRef] [PubMed]
62.
Snyder, G.A.; Deredge, D.; Waldhuber, A.; Fresquez, T.; Wilkins, D.Z.; Smith, P.T.; Durr, S.; Cirl, C.; Jiang, J.; Jennings, W.; et al.
Crystal structures of the Toll/Interleukin-1 receptor (TIR) domains from the Brucella protein TcpB and host adaptor TIRAP reveal
mechanisms of molecular mimicry. J. Biol. Chem. 2014,289, 669–679. [CrossRef]
63.
Durward, M.; Radhakrishnan, G.; Harms, J.; Bareiss, C.; Magnani, D.; Splitter, G.A. Active evasion of CTL mediated killing and
low quality responding CD8+ T cells contribute to persistence of Brucellosis. PLoS ONE 2012,7, e34925. [CrossRef] [PubMed]
64.
Lopez-Santiago, R.; Sanchez-Argaez, A.B.; De Alba-Nunez, L.G.; Baltierra-Uribe, S.L.; Moreno-Lafont, M.C. Immune Response to
Mucosal Brucella Infection. Front. Immunol. 2019,10, 1759. [CrossRef] [PubMed]
65.
de Jong, M.F.; Rolan, H.G.; Tsolis, R.M. Innate immune encounters of the (Type) 4th kind: Brucella.Cell Microbiol.
2010
,12,
1195–1202. [CrossRef]
66.
Hong, P.C.; Tsolis, R.M.; Ficht, T.A. Identification of genes required for chronic persistence of Brucella abortus in mice. Infect.
Immun. 2000,68, 4102–4107. [CrossRef]
67. Percin, D. Microbiology of Brucella. Recent Pat. Antiinfect. Drug Discov. 2013,8, 13–17. [CrossRef]
Int. J. Mol. Sci. 2021,22, 3673 13 of 13
68.
Barbier, T.; Zuniga-Ripa, A.; Moussa, S.; Plovier, H.; Sternon, J.F.; Lazaro-Anton, L.; Conde-Alvarez, R.; De Bolle, X.; Iriarte, M.;
Moriyon, I.; et al. Brucella central carbon metabolism: An update. Crit. Rev. Microbiol. 2018,44, 182–211. [CrossRef]
69.
Fretin, D.; Fauconnier, A.; Kohler, S.; Halling, S.; Leonard, S.; Nijskens, C.; Ferooz, J.; Lestrate, P.; Delrue, R.M.; Danese, I.; et al.
The sheathed flagellum of Brucella melitensis is involved in persistence in a murine model of infection. Cell Microbiol.
2005
,7,
687–698. [CrossRef]
70.
Barrionuevo, P.; Giambartolomei, G.H. Inhibition of antigen presentation by Brucella: Many more than many ways. Microbes.
Infect. 2019,21, 136–142. [CrossRef] [PubMed]
71. Glass, C.K.; Natoli, G. Molecular control of activation and priming in macrophages. Nat. Immunol. 2016,17, 26–33. [CrossRef]
72.
Delpino, M.V.; Barrionuevo, P.; Macedo, G.C.; Oliveira, S.C.; Genaro, S.D.; Scian, R.; Miraglia, M.C.; Fossati, C.A.; Baldi, P.C.;
Giambartolomei, G.H. Macrophage-elicited osteoclastogenesis in response to Brucella abortus infection requires TLR2/MyD88-
dependent TNF-alpha production. J. Leukoc. Biol. 2012,91, 285–298. [CrossRef]
73.
Fernandez-Lago, L.; Monte, M.; Chordi, A. Endogenous gamma interferon and interleukin-10 in Brucella abortus 2308 infection in
mice. FEMS Immunol. Med. Microbiol. 1996,15, 109–114. [CrossRef]
74.
Fernandes, D.M.; Jiang, X.; Jung, J.H.; Baldwin, C.L. Comparison of T cell cytokines in resistant and susceptible mice infected
with virulent Brucella abortus strain 2308. FEMS Immunol. Med. Microbiol. 1996,16, 193–203. [CrossRef]
75.
Fernandes, D.M.; Baldwin, C.L. Interleukin-10 downregulates protective immunity to Brucella abortus.Infect. Immun.
1995
,63,
1130–1133. [CrossRef]
76.
Xavier, M.N.; Winter, M.G.; Spees, A.M.; Nguyen, K.; Atluri, V.L.; Silva, T.M.; Baumler, A.J.; Muller, W.; Santos, R.L.; Tsolis,
R.M. CD4+ T cell-derived IL-10 promotes Brucella abortus persistence via modulation of macrophage function. PLoS Pathog
2013
,
9, e1003454. [CrossRef] [PubMed]
77.
Spera, J.M.; Ugalde, J.E.; Mucci, J.; Comerci, D.J.; Ugalde, R.A. A B lymphocyte mitogen is a Brucella abortus virulence factor
required for persistent infection. Proc. Natl. Acad. Sci. USA 2006,103, 16514–16519. [CrossRef]
78.
Kim, S.; Watanabe, K.; Suzuki, H.; Watarai, M. Roles ofBrucella abortus SpoT in morphological differentiation and intramacrophagic
replication. Microbiology (Reading) 2005,151, 1607–1617. [CrossRef]
79.
Salcedo, S.P.; Chevrier, N.; Lacerda, T.L.S.; Ben Amara, A.; Gerart, S.; Gorvel, V.A.; de Chastellier, C.; Blasco, J.M.; Mege, J.L.;
Gorvel, J.P. Pathogenic Brucella replicate in human trophoblasts. J. Infect. Dis. 2013,207, 1075–1083. [CrossRef] [PubMed]
80.
Anderson, T.D.; Meador, V.P.; Cheville, N.F. Pathogenesis of placentitis in the goat inoculated with Brucella abortus. I. Gross and
histologic lesions. Vet. Pathol. 1986,23, 219–226. [CrossRef] [PubMed]
81.
Meador, V.P.; Deyoe, B.L. Intracellular localization of Brucella abortus in bovine placenta. Vet. Pathol.
1989
,26, 513–515. [CrossRef]
[PubMed]
... Internalization of smooth or virulent Brucella species occurs through the lipid rafts present on the macrophage cell. On the other hand, internalization of rough or avirulent species does not occur through lipid rafts but normally by the process of phagocytosis hence they get connected rapidly with the lysosome resulting destruction of the bacteria which shows that lipid raft is a necessary condition for the survival of early stage of bacteria in the host cell [78,79]. ...
... Moreover, brucellar T4SS plays an important role in keeping away the BCVs from the host immune response. The early BCVs now interact with the endoplasmic reticulum (ER) to form replicative BCVs (rBCV) which are characterized by several ER markers, namely calnexin, calreticulin, and Sec61β (Figure 4B) [79]. Bacterial virulence factor, cyclic β-1-2-glucans (CβG) assists BCVs in combining with ER. ...
Chapter
Full-text available
In 2020-2022, infectious diseases were the major causes of infection and death globally. Many viral and bacterial diseases are starting to emerge/reemerge frequently. The zoonotic infections were reported to be dominant in a few decades. Now the holis-tic one-health approach is the need of the hour to tackle the emerging and reemerging pathogens by extensive and heedless use of antimicrobials, lack of novel antimicrobi-als, and unavailability of appropriate vaccines increased the severity of pathogens. Brucellosis is a well-known zoonotic disease also called undulant fever, Malta fever, Mediterranean fever, etc. Many components of the genus Brucella will be discussed in this chapter, namely pathogenesis, genes/proteins responsible for diseases in animal and humans, available treatment options, drug resistance phenomenon, etc.
... The ER provides an optimal environment for Brucella to replicate and enhance its pathogenicity (Sedzicki et al., 2018). Current studies suggest that eBCV can merge with the ER via COP II vesicles and activate Sar1 protein during vesicle transport (Celli, 2019;Jiao et al., 2021). Additionally, the fusion of eBCV with the ER is dependent on the small GTPase Rab2 (Fugier et al., 2009). ...
... In later stages of infection, rBCV transforms into autophagic Brucella-containing vacuoles (aBCV), which differ from traditional autophagosomes. Intriguingly, the formation of aBCV requires autophagy initiation factors like ULK1, Beclin1, ATG14L, and PI3K kinase, but it does not involve autophagy extension factors such as ATG5, ATG16L1, ATG4B, ATG7, and LC3 (Jiao et al., 2021;Starr et al., 2012). Eventually, Brucella completes its intracellular cycle and is released through lytic and non-lytic mechanisms, allowing for a new round of infection ( Fig. 1). ...
Article
Full-text available
Brucella, an adept intracellular pathogen, causes brucellosis, a zoonotic disease leading to significant global impacts on animal welfare and the economy. Regrettably, there is currently no approved and effective vaccine for human use. The ability of Brucella to evade host defenses is essential for establishing chronic infection and ensuring stable intracellular growth. Brucella employs various mechanisms to evade and undermine the innate and adaptive immune responses of the host through modulating the activation of pattern recognition receptors (PRRs), inflammatory responses, or the activation of immune cells like dendritic cells (DCs) to inhibit antigen presentation. Moreover, it regulates multiple cellular processes such as apoptosis, pyroptosis, and autophagy to establish persistent infection within host cells. This review summarizes the recently discovered mechanisms employed by Brucella to subvert host immune responses and research progress on vaccines, with the aim of advancing our understanding of brucellosis and facilitating the development of more effective vaccines and therapeutic approaches against Brucella.
... The organism known as Brucella is devoid of classical virulence factors such exotoxins, pili, endotoxins, flagella, and plasmids (Głowacka et al., 2018). The capacity of Brucella to prevent lysosomes from fusing with phagosomes, which leads to degranulation and the activation of the myelo-peroxidase-halide system, as well as to prevent tumor necrosis factors and cell death in host cells, is linked to its ability to survive and multiply within host cells after evading the host's defense mechanisms (Jiao et al., 2021). Malignant Brucella species can infect and live in both phagocytic and non-phagocytic phagocytes, such as macrophages (Huy et al., 2022). ...
Article
Full-text available
One zoonotic infectious animal disease is brucellosis. The bacteria that cause brucellosis belong to the genus Brucella. Numerous animal and human species are affected by Brucellosis, with an estimated 500,000 human cases recorded annually worldwide. The occurrence of new areas of infection and the resurgence of infection in already infected areas indicate how dynamically Brucellosis is distributed throughout different geographic regions. Bacteria originate from the blood and are found in the reticuloendothelial system, the liver, the spleen, and numerous other locations, including the joints, kidneys, heart, and genital tract. Diagnosis of this disease can be done by Bacterial Isolation, Molecular tests, Modified acid-fast Stain, Rose Bengal Test (RBT), Milk Ring Test (MRT), Complement Fixation Test (CFT), Enzyme-Linked Immunosorbent Assay (ELISA), and Serum Agglutination Test (SAT). The primary sign of a B. abortus infection is infertility, which can result in abortion and the birth of a frail fetus that may go on to infect other animals. In humans, the main symptoms are acute febrile illness, with or without localization signs, and chronic infection. Female cattle have a greater risk of contracting Brucella disease. Human populations at high risk of contracting brucellosis include those who care for cattle, veterinarians, slaughterhouse employees, and butchers. Antibiotic treatment of brucellosis is often unsuccessful due to the intracellular survival of Brucella and its adaptability in macrophages. A 'One Health' strategy is necessary to control illnesses like brucellosis.
... It is well known that Brucella has developed a wide range of strategies to evade both innate and adaptive immune responses (Martirosyan and Gorvel, 2013;Skendros and Boura, 2013;Jiao et al., 2021). The main escape mechanisms of Brucella against the host immune system are inhibition of the complement pathway and TLR signaling pathways, interference with antigen presentation, selective subversion of the autophagy pathway, inhibition of DC stimulation, inhibition of autophagic lysosomal fusion, and macrophage apoptosis ( ...
Article
Full-text available
Brucella consists of gram-negative bacteria that have the ability to invade and replicate in professional and non-professional phagocytes, and its prolonged persistence in the host leads to brucellosis, a serious zoonosis. Toll-like receptors (TLRs) are the best-known sensors of microorganisms implicated in the regulation of innate and adaptive immunity. In particular, TLRs are transmembrane proteins with a typical structure of an extracellular leucine-rich repeat (LRR) region and an intracellular Toll/interleukin-1 receptor (TIR) domain. In this review, we discuss Brucella infection and the aspects of host immune responses induced by pathogens. Furthermore, we summarize the roles of TLRs in Brucella infection, with substantial emphasis on the molecular insights into its mechanisms of action.
... They can be categorized as obligate, unable to multiply outside host cells, or facultative, with the ability to multiply both inside and outside cells. Infections caused by intracellular bacteria include brucellosis, listeriosis, tuberculosis, and salmonellosis (Silva, 2012;Jiao et al., 2021). These pathogens use various pathways to enter host cells, such as macrophages, phagocytes, epithelial and endothelial cells, and hepatocyte (Kaufmann, 1993;Thakur et al., 2019). ...
Article
Full-text available
Bacteria are the most prevalent form of microorganisms and are classified into two categories based on their mode of existence: intracellular and extracellular. While most bacteria are beneficial to human health, others are pathogenic and can cause mild to severe infections. These bacteria use various mechanisms to evade host immunity and cause diseases in humans. The susceptibility of a host to bacterial infection depends on the effectiveness of the immune system, overall health, and genetic factors. Malnutrition, chronic illnesses, and age-related vulnerabilities are the additional confounders to disease severity phenotypes. The impact of bacterial pathogens on public health includes the transmission of these pathogens from healthcare facilities, which contributes to increased morbidity and mortality. To identify the most significant threats to public health, it is crucial to understand the global burden of common bacterial pathogens and their pathogenicity. This knowledge is required to improve immunization rates, improve the effectiveness of vaccines, and consider the impact of antimicrobial resistance when assessing the situation. Many bacteria have developed antimicrobial resistance, which has significant implications for infectious diseases and favors the survival of resilient microorganisms. This review emphasizes the significance of understanding the bacterial pathogens that cause this health threat on a global scale.
... Brucellosis is caused by Gram-negative bacteria of the genus Brucella, which have the ability to establish longterm infections in their hosts [6,7]. Brucella evades the host immune system's clearance mechanisms, and the duration of infection within host cells depends on the host's immunity and the appropriate use of specific antibiotics. ...
Article
Full-text available
Background Brucellosis is a severe zoonotic disease that is often overlooked, particularly in impoverished countries. Timely identification of focal complications in brucellosis is crucial for improving treatment outcomes. However, there is currently a lack of established indicators or biomarkers for diagnosing these complications. Therefore, this study aimed to investigate potential warning signs of focal complications in human brucellosis, with the goal of providing practical parameters for clinicians to aid in the diagnosis and management of patients. Methods A multi-center cross-sectional study was conducted in China from December 2019 to August 2021. The study aimed to investigate the clinical characteristics and complications of patients with brucellosis using a questionnaire survey and medical record system. The presence of warning signs for complications was assessed using univariate and multivariate logistic regression models. Receiver operating characteristic (ROC) curves and the area under the curve (AUC) were used for variable screening and model evaluation. Results A total of 880 participants diagnosed with human brucellosis were enrolled. The median age of the patients was 50 years [interquartile range ( IQR ): 41.5–58.0], and 54.8% had complications. The most common organ system affected by complications was the osteoarticular system (43.1%), with peripheral arthritis (30.0%), spondylitis (16.6%), paravertebral abscess (5.0%), and sacroiliitis (2.7%) being the most prevalent. Complications in other organ systems included the genitourinary system (4.7%), respiratory system (4.7%), and hematologic system (4.6%). Several factors were found to be associated with focal brucellosis. These factors included a long delay in diagnosis [odds ratio ( OR ) = 3.963, 95% confidence interval ( CI ) 1.906–8.238 for > 90 days], the presence of underlying disease ( OR = 1.675, 95% CI 1.176–2.384), arthralgia ( OR = 3.197, 95% CI 1.986–5.148), eye bulging pain ( OR = 3.482, 95% CI 1.349–8.988), C-reactive protein (CRP) > 10 mg/L ( OR = 1.910, 95% CI 1.310–2.784) and erythrocyte sedimentation rate (ESR) elevation ( OR = 1.663, 95% CI 1.145–2.415). The optimal cutoff value in ROC analysis was > 5.4 mg/L for CRP (sensitivity 73.4% and specificity 51.9%) and > 25 mm/h for ESR (sensitivity 47.9% and specificity 71.1%). Conclusions More than 50% of patients with brucellosis experienced complications. Factors such as diagnostic delay, underlying disease, arthralgia, eye pain, and elevated levels of CRP and ESR were identified as significant markers for the development of complications. Therefore, patients presenting with these conditions should be closely monitored for potential complications, regardless of their culture results and standard tube agglutination test titers.
... However, when stimulated, NF-κB is liberated from its inhibitor and migrates into the nucleus to initiate various functions, including the regulation of cytokines, such as IL-6, IL-10, and TNF-α [27]. The activation of NF-κB is crucial for mounting an effective immune response against Brucella invasion [28,29]. ...
Article
Full-text available
Catalase, an antioxidant enzyme widely produced in mammalian cells and bacteria, is crucial to mitigating oxidative stress in hostile environments. This function enhances the intracellular survivability of various intracellular growth pathogens, including Brucella (B.) abortus. In this study, to determine whether the suppression of catalase can inhibit the intracellular growth of B. abortus, we employed 3-amino-1,2,4-triazole (3-AT), a catalase inhibitor, in both RAW 264.7 macrophage cells and an ICR mouse model during Brucella infection. The intracellular growth assay indicated that 3-AT exerts growth-inhibitory effects on B. abortus within macrophages. Moreover, it contributes to the accumulation of reactive oxygen species and the formation of nitric oxide. Notably, 3-AT diminishes the activation of the nucleus transcription factor (NF-κB) and modulates the cytokine secretion within infected cells. In our mouse model, the administration of 3-AT reduced the B. abortus proliferation within the spleens and livers of infected mice. This reduction was accompanied by a diminished immune response to infection, as indicated by the lowered levels of TNF-α, IL-6, and IL-10 and altered CD4+/CD8+ T-cell ratio. These results suggest the protective and immunomodulatory effects of 3-AT treatment against Brucella infection.
... Brucellosis is caused by facultative intracellular Gram-negative bacteria of the genus Brucella, which are able to establish long-term infections in their hosts [5,6]. Brucella can evade the clearance mechanisms of the host immune system. ...
Preprint
Full-text available
Background Brucellosis is one of the most serious “ignored zoonoses” in the world and especially affects poverty-stricken nations. Early diagnosis of focal complications in brucellosis increases treatment success. However, up to now, no clear joint indicators of symptoms or biomarkers for the diagnosis of complications. Hence, we investigated the warning signs of focal complications in human brucellosis aimed to provide practical parameters for clinicians in patients diagnosis and management. Methods A multi‑center cross‑sectional study was conducted in China from December 2019 to August 2021. The clinical characteristics and complications of brucellosis patients were based on questionnaire survey and medical record system. Warning signs of complications were assessed by univariate and multivariate logistic regression models. Receiver operating characteristic (ROC) curves and the area under the curve (AUC) were used for variable screening and model evaluation. Results A total of 880 participants of human brucellosis were enrolled. Of the patients, median age was 50 years [interquartile range (IQR):41.5,58.0] and 54.8% had complications. The complication involved any organ, common in osteoarticular system (43.1%), including peripheral arthritis (30.0%), spondylitis (16.6%), paravertebral abscess (5.0%) and sacroiliitis (2.7%); followed by genitourinary system (4.7%), respiratory system (4.7%), and hematologic system (4.6%). Long delay in diagnosis [odds ratio (OR):3.963, 95% confidence interval (CI):1.906–8.238 for > 90days], underlying disease(OR:1.675, 95%CI:1.176–2.384), arthralgia(OR:3.197, 95%CI:1.986–5.148), bulging pain(OR:3.482, 95%CI:1.349–8.988), C-reactive protein (CRP) > 10 mg/L (OR:1.910, 95%CI:1.310–2.784)and erythrocyte sedimentation rate (ESR) elevation (OR:1.663, 95%CI:1.145–2.415)were the relevant factors for focal brucellosis. The optimal cutoff value in ROC analysis was > 5.4 mg/L for CRP (sensitivity 73.4% and specificity 51.9%) and > 25 mm/h for ESR (sensitivity 47.9% and specificity 71.1%). Conclusions Over half of brucellosis patients suffered complications. Diagnostic delay, underlying disease, arthralgia, bulging pain, and high CRP and ESR were significant relevant markers of complications. Regardless of culture results and standard tube agglutination test (SAT) titers, patients with these conditions should be on the alert for complications.
Article
Full-text available
Brucella is a zoonotic intracellular bacterium that poses threats to human health and economic security. Intracellular infection is a hallmark of the agent Brucella and a primary cause of distress, through which the bacterium regulates the host intracellular environment to promote its own colonization and replication, evading host immunity and pharmaceutical killing. Current studies of Brucella intracellular processes are typically premised on bacterial phenotype such as intracellular bacterial survival, followed by biochemical or molecular biological approaches to reveal detailed mechanisms. While such processes can deepen the understanding of Brucella -host interaction, the insights into host alterations in infection would be easily restricted to known pathways. In the current study, we applied CRISPR Cas9 screen to identify host genes that are most affected by Brucella infection on cell viability at the genomic level. As a result of CRISPR screening, we firstly identified that knockout of the negatively selected genes GOLGA6L6 , DEFB103B , OR4F29 , and ERCC6 attenuate the viability of both the host cells and intracellular Brucella , suggesting these genes to be potential therapeutic targets for Brucella control. In particular, knockout of DEFB103B diminished Brucella intracellular survival by altering host cell autophagy. Conversely, knockout of positive screening genes promoted intracellular proliferation of Brucella . In summary, we screened host genes at the genomic level throughout Brucella infection, identified host genes that are previously not recognized to be involved in Brucella infection, and provided targets for intracellular infection control. IMPORTANCE Brucella is a Gram-negative bacterium that infects common mammals causing arthritis, myalgia, neuritis, orchitis, or miscarriage and is difficult to cure with antibiotics due to its intracellular parasitism. Therefore, unraveling the mechanism of Brucella -host interactions will help controlling Brucella infections. CRISPR-Cas9 is a gene editing technology that directs knockout of individual target genes by guided RNA, from which genome-wide gene-knockout cell libraries can be constructed. Upon infection with Brucella , the cell library would show differences in viability as a result of the knockout and specific genes could be revealed by genomic DNA sequencing. As a result, genes affecting cell viability during Brucella infection were identified. Further testing of gene function may reveal the mechanisms of Brucella -host interactions, thereby contributing to clinical therapy.
Article
Full-text available
The results of study relationship between antigen reactivity of T-lymphocyte population under ex vivo conditions and the intensity of protective post-vaccination immunity to causative agent of brucellosis are presented. Тaking into account the peculiarities of immunopathogenesis brucellosis and prevailing role of adaptive T-cell immunity to protect against the causative agent of infection, possibility predictive evaluation of protective immunity against brucellosis using CAST-tests is considered as the most important aspect of brucellosis problems. There is an obvious need for an ex vivo correlation analysis of the activity of antigen stimulation of T cells and the intensity of protective immunity formed after vaccination. A close direct proportional relationship was established between the number of live microbial cells Brucella abortus 19BA vaccine strain administered and increase in ex vivo CD3-cell activation. A close correlation was revealed between ex vivo value of antigen-induced stimulation CD3-lymphocytes and level of post-vaccination immunological protection against brucellosis infection. It has been shown that in biomodels vaccinated against brucellosis with a T-lymphocyte stimulation coefficient of 50% or more (according to intensity of antigen-induced ex vivo expression CD25), 100% protection from the development of brucellosis infection after infection with Brucella melitensis at a dose of 1 × 10 ³ live microbial cells are provided. At the same time, there was a lack of a close correlation between an increase in the dose of brucella vaccine strain administered to biomodels and a change in geometric mean antibody titer, presence of a weakly pronounced relationship between level of agglutinins and immunological protection of biomodels from development brucellosis infection and indicators bacterial contamination body.Based on results of study, it was demonstrated that it is possible to quantify the formation and protective activity of T-cell immunity to causative agent of brucellosis based on analysis of level antigen reactivity of CD3-lymphocytes ex vivo . The data obtained and described methodological approach can be used as a predictive criterion in assessing protective level of cellular immunity to causative agent of brucellosis in vaccinated or recovering patients, as well as in order to analyze effectiveness of specific prophylaxis brucellosis and study immunogenicity and protective properties candidate for brucellosis vWe present the results of studies related to antigen reactivity of T lymphocyte population under ex vivo conditions and the intensity of protective post-vaccination immunity to causative agent of brucellosis. Due to peculiarities of immunopathogenesis in brucellosis infection and prevailing role of adaptive T cell immunity for protection against the causative agent of infection, a predictive evaluation of protective immunity against brucellosis using CAST-tests is considered the most important issue in the field. There is an obvious need for ex vivo analysis of correlations between the activity of antigen stimulation of T cells, and the intensity of protective immunity raised after vaccination. A close direct relationship was established between the number of live microbial cells of Brucella abortus 19BA vaccine strain administered, and increase in ex vivo CD3 cell activation. A close correlation (r = -0.841 ÷ -0.966, R2 = 0.708 ÷ 0.969) was revealed between ex vivo values of antigeninduced stimulation of CD3 lymphocytes, and the levels of post-vaccination immunological protection against brucellosis infection. We have shown that, in biomodels vaccinated against brucellosis with a T lymphocyte stimulation coefficient of 50% or more (according to intensity of antigen-induced ex vivo CD25 expression), 100% protection against brucellosis infection was achieved after contamination with B rucella melitensis at a dose of 1×10 ³ live microbial cells. At the same time, a lack of a close correlation was noted between an increased dose of Brucella vaccine strain administered to biomodels, and a change in geometric mean of antibody titer ( R2 = 0.357÷0.404), along with a weak relationship between the levels of agglutinins and immunological protection of biomodels from developing brucellosis infection and indices of in vivo bacterial contamination.These results suggest an opportunity to quantify development and protective activity of T cell immunity to the causal agent of brucellosis based ex vivo levels of antigen reactivity of CD3 lymphocytes. A correlation analysis between the state of T cell antigen reactivity and immunological resistance to brucellosis infection indicated a high degree of closeness between these indices. The key influence on activity of protective immunity is exerted by the levels of antigen reactivity of T lymphocytes, whereas the quotient of antigenic stimulation in CD3+CD25+ population may be considered the most informative index of immune protective activity. The data obtained and the described methodology may be used as a predictive criterion in assessing protective level of cellular immunity to causative agent of brucellosis in vaccinated or recovering patients, testing the efficiency of specific prophylaxis in brucellosis and studying immunogenicity and protective properties of candidate vaccines against brucellosis.
Article
Full-text available
Brucellosis is one of the most prevalent bacterial zoonosis of worldwide distribution. The disease is caused by Brucella spp., facultative intracellular pathogens. Brucellosis in animals results in abortion of fetuses, while in humans, it frequently manifests flu-like symptoms and a typical undulant fever, being osteoarthritis a common complication of the chronic infection. The two most common ways to acquire the infection in humans are through the ingestion of contaminated dairy products or by inhalation of contaminated aerosols. Brucella spp. enter the body mainly through the gastrointestinal and respiratory mucosa; however, most studies of immune response to Brucella spp. are performed analyzing models of systemic immunity. It is necessary to better understand the mucosal immune response induced by Brucella infection since this is the main entry site for the bacterium. In this review, some virulence factors and the mechanisms needed for pathogen invasion and persistence are discussed. Furthermore, some aspects of local immune responses induced during Brucella infection will be reviewed. With this knowledge, better vaccines can be designed focused on inducing protective mucosal immune response.
Article
Full-text available
The Intracellular Life Cycle of Brucella spp., Page 1 of 2 Abstract Bacteria of the genus Brucella colonize a wide variety of mammalian hosts, in which their infectious cycle and ability to cause disease predominantly rely on an intracellular lifestyle within phagocytes. Upon entry into host cells, Brucella organisms undergo a complex, multistage intracellular cycle in which they sequentially traffic through, and exploit functions of, the endocytic, secretory, and autophagic compartments via type IV secretion system (T4SS)-mediated delivery of bacterial effectors. These effectors modulate an array of host functions and machineries to first promote conversion of the initial endosome-like Brucella-containing vacuole (eBCV) into a replication-permissive organelle derived from the host endoplasmic reticulum (rBCV) and then to an autophagy-related vacuole (aBCV) that mediates bacterial egress. Here we detail and discuss our current knowledge of cellular and molecular events of the Brucella intracellular cycle. We discuss the importance of the endosomal stage in determining T4SS competency, the roles of autophagy in rBCV biogenesis and aBCV formation, and T4SS-driven mechanisms of modulation of host secretory traffic in rBCV biogenesis and bacterial egress.
Article
Full-text available
Brucella organisms are intracellular stealth pathogens of animals and humans. These bacteria overcome the assault of innate immunity at early stages of the infection. Removal of neutrophils (PMNs) at the onset of adaptive immunity against Brucella abortus , favored the bacterial elimination in mice. This was associated with higher levels of IFN-γ, and higher proportion of cells expressing IL-6 and iNOS, compatible with M1 macrophages, in the PMN-depleted B. abortus infected mice (PMNd- Br- mice). At later times of the acute infection phase, the amounts of IFN-γ lowered while the IL-6, IL-10 and IL-12 became the predominant cytokines in the PMNd- Br- mice. IL-4, IL-1β and TNF-α remained at background levels at all times of the infection. Depletion of PMNs at the acute stages of infection promoted the premature resolution of spleen inflammation. The efficient removal of bacteria in the PMNd- Br- mice was not due to the increase of antibodies, since the immunoglobulin isotype responses to Brucella antigens were dampen. Anti- Brucella antibodies abrogated the production of IL-6, IL-10 and IL-12 but did not affect the levels of IFN-γ at later stages of infection in the PMNd- Br- mice. These results demonstrate that PMNs have an active role in modulating the course of B. abortus infection, after the adaptive immune response has already developed.
Article
Full-text available
As dendritic cells (DCs) are among the first cells to encounter antigens, these cells trigger both innate and T cell responses, and are the most potent antigen-presenting cells. Brucella spp., which is an intracellular facultative and stealthy pathogen, is able to evade the bactericidal activities of professional phagocytes. Several studies have demonstrated that Brucella can survive and replicate intracellularly, thereby provoking impaired maturation of DCs. Therefore, the interaction between DCs and Brucella becomes an interesting model to study the immune response. In this review, we first will describe the most common techniques for DCs differentiation in vitro as well as general features of brucellosis. Then, the interaction of DCs and Brucella, including pathogen recognition, molecular mechanisms of bacterial pathogenesis, and intracellular trafficking of Brucella to subvert innate response, will be reviewed. Finally, we will debate diversity in immunological DC response and the controversial role of DC activation against Brucella infection.
Article
Full-text available
Brucellaceae are a group of pathogenic intracellular bacteria with the ability to modulate the host response, both at the individual cell level and systemically. One of the hallmarks of the virulence process is the capacity of the bacteria to downregulate the adaptive and acquired host immune response through a plethora of virulence factors that directly impact several key signaling cascades. PrpA is one of those virulence factors that alters, via its polyclonal B-cell activity, the humoral and cellular immune responses of the host, ultimately favoring the establishment of a chronic infection. Even though PrpA affects B-cells, it directly targets macrophages, triggering a response that ultimately affects B-lymphocytes. In the present manuscript we report that PrpA is S-palmitoylated in two N-terminal cysteine residues by the host cell and that this modification is necessary for its biological activity. Our results demonstrate that S-palmitoylation promotes PrpA migration to the host cell plasma membrane and stabilizes the protein during infection. These findings add a new mechanism exploited by this highly evolved pathogen to modulate the host immune response.
Article
Full-text available
Brucellae are Gram-negative, small rods infecting mammals and capable of causing disease called brucellosis. The infection results in abortion and sterility in domestic animals (sheeps, pigs, rams etc). Especially dangerous for humans are: Brucella melitensis, Brucella suis, Brucella abortus, and Brucella canis that trigger unspecific symptoms (flu-like manifestation). Brucella rods are introduced via host cells, by inhalation, skin abrasions, ingestion or mucosal membranes. The most important feature of Brucella is the ability to survive and multiply within both phagocytic and non-phagocytic cells. Brucella does not produce classical virulence factors: exotoxin, cytolisins, exoenzymes, plasmids, fimbria, and drug resistant forms. Major virulence factors are: lipopolysaccharide (LPS), T4SS secretion system and BvrR/BvrS system, which allow interaction with host cell surface, formation of an early, late BCV (Brucella Containing Vacuole) and interaction with endoplasmic reticulum (ER) when the bacteria multiply. The treatment of brucellosis is based on two-drug therapy, the most common combinations of antibiotics are: doxycycline with rifampicin or fluoroquinolones with rifampicin. Currently, also other methods are used to disrupt Brucella intracellular replication (tauroursodeoxycholic acid or ginseng saponin fraction A).
Article
Full-text available
The lipopolysaccharide (LPS) is a major virulence factor of Brucella, a facultative intracellular pathogenic Gram-negative bacterium. Brucella LPS exhibits a low toxicity and its atypical structure was postulated to delay the host immune response, favouring the establishment of chronic disease. Here we carried out an in-depth in vitro and in vivo characterisation of the immunomodulatory effects of Brucella LPS on different dendritic cell (DC) subpopulations. By using LPSs from bacteria that share some of Brucella LPS structural features, we demonstrated that the core component of B. melitensis wild-type (Bm-wt) LPS accounts for the low activation potential of Brucella LPS in mouse GM-CSF-derived (GM-) DCs. Contrary to the accepted dogma considering Brucella LPS a poor TLR4 agonist and DC activator, Bm-wt LPS selectively induced expression of surface activation markers and cytokine secretion from Flt3-Ligand-derived (FL-) DCs in a TLR4-dependent manner. It also primed in vitro T cell proliferation by FL-DCs. In contrast, modified LPS with a defective core purified from Brucella carrying a mutated wadC gene (Bm-wadC), efficiently potentiated mouse and human DC activation and T cell proliferation in vitro. In vivo, Bm-wt LPS promoted scant activation of splenic DC subsets and limited recruitment of monocyte- DC like cells in the spleen, conversely to Bm-wadC LPS. Bm-wadC live bacteria drove high cytokine secretion levels in sera of infected mice. Altogether, these results illustrate the immunomodulatory properties of Brucella LPS and the enhanced DC activation ability of the wadC mutation with potential for vaccine development targeting Brucella core LPS structure.
Article
Full-text available
The brucellae are facultative intracellular pathogens causing brucellosis, an important zoonosis. Here, we review the nutritional, genetic, proteomic and transcriptomic studies on Brucella carbon uptake and central metabolism, information that is needed for a better understanding of Brucella virulence. There is no uniform picture across species but the studies suggest primary and/or secondary transporters for unknown carbohydrates, lactate, glycerol phosphate, erythritol, xylose, ribose, glucose and glucose/galactose, and routes for their incorporation to central metabolism, including an erythritol pathway feeding the pentose phosphate cycle. Significantly, all brucellae lack phosphoenolpyruvate synthase and phosphofructokinase genes, which confirms previous evidence on glycolysis absence, but carry all Entner–Doudoroff (ED) pathway and Krebs cycle (and glyoxylate pathway) genes. However, glucose catabolism proceeds through the pentose phosphate cycle in the classical species, and the ED pathway operates in some rodent-associated brucellae, suggesting an ancestral character for this pathway in this group. Gluconeogenesis is functional but does not rely exclusively on classical fructose bisphosphatases. Evidence obtained using infection models is fragmentary but suggests the combined or sequential use of hexoses/pentoses, amino acids and gluconeogenic substrates. We also discuss the role of the phosphotransferase system, stringent reponse, quorum sensing, BvrR/S and sRNAs in metabolism control, an essential aspect of the life style of facultative intracellular parasites.
Article
Brucella infection activates the immune system and favors the differentiation of CD4⁺ and CD8⁺ T cells. To persist during a long time inside macrophages evading immune surveillance of these T cells the pathogen must exploit different evasion strategies. We review the mechanisms whereby Brucella, through TLR signaling, inhibits MHC class I and II antigen presentation, allowing infected macrophages to become effective niches for Brucella survival.
Article
Brucella abortus can survive and replicate within host macrophages, and great efforts have been made to demonstrate the genes involved in pathogenicity such as internalization in Brucella research. Therefore, intracellular responses were compared between THP-1 macrophage cells stimulated with B. abortus wild-type and four mutants (C1, C10, C27, and C32) using microarray to demonstrate the role of genes related to intracellular survival and replication. These mutants were generated by deleting genes encoding BAB_RS13225 (4-hydrobenzoate 3-monooxygenase, PHBH), BAB_RS00455 (heme exporter protein cytochrome C, CcmC), BAB_RS03675 (exopolyphosphatase, PPX), and BAB_RS13225 (peptidase M24). The result showed that mutants C1 and C10 induced significant suppression of survival levels and cytokine expression relative to wild-type in the THP-1 macrophage cells. These findings suggest that the BAB_RS13225 and BAB_RS00455 genes play important roles in survival within human macrophages. Conversely, mutants C27 and C32 induced significantly higher survival level than wild-type in the cells inhibiting cellular signal transduction. It is assumed that BAB_RS03675 and BAB_RS13225 genes play a role in cellular resistance to B. abortus. Therefore, the disrupted genes are involved in B. abortus intracellular growth, especially survival, and they could be effective targets for understanding the intracellular bacteria, B. abortus.