Transcript Case_4x

CASE 4
THE IMMUNE RESPONSE
Ashley Wang
Path417
March 2016
The Case
10-year-old Ronnie McDonald has developed abdominal
cramps, bloody diarrhea and a low grade fever. His parents
take him to see the family doctor. The doctor asks about what
Ronnie may have eaten in the past week and his parents recall
that last weekend at a neighbor’s barbecue they were
concerned that the hamburgers may not have been cooked
thoroughly and Ronnie had eaten two burgers. The doctor
performs a physical examination noting no rebound tenderness
just some mild periumbilical tenderness. He asks the parents to
collect a stool sample for the Microbiology Laboratory and to
take Ronnie to the local lab for some routine bloodwork.
The Cause
Foodborne illness-causing organisms, include but are not limited to
Organism
Onset Time
Signs & Symptoms
Duration
Food Source
Salmonella
6 – 48 hrs
Diarrhea, fever,
abdominal cramps,
vomiting
4 – 7 days
Eggs, poultry,
meat…
Shigella
4 – 7 days
Abdominal cramps,
fever, and diarrhea.
Stools may contain
blood and mucous
24 – 48 hrs
Raw produce,
uncooked
foods,
contaminated
water…
Although the onset time is not provided, Ronnie is more likely to
be infected with Salmonella based on the epidemiology.
Q1: Host Response
What elements of the innate and adaptive
(humoural and cellular) immune response are
involved in this infection.
Basic Anatomy of The GI Tract






Gastrointestinal Barrier
A single layer of intestinal epithelial cells (IEC) that provides a physical
and chemical barrier to the gut
IEC express TLR5 on basolateral side, low levels of TLR2 and TLR4 on
apical side, and NOD1/NOD2 inside the cells
Tight junctions between these cells to block the entry of pathogen and
commensals
Lamina propria
Houses a variety of lymphocytes, including T cells, B cells, dendritic cells,
macrophages, and neutrophils

Contains Peyer’s patches (PP), which are aggregated lymphoid follicles

M cells are specialized cells that transport bacteria via transcytosis
Broz, P., et. al,
2012
Fig. 1 The Gastrointestinal Immune System
Schematic representation of the structure of immune system in the gastrointestinal
tract and the different ways Salmonella can take to invade the intestinal mucosa.
Initial Barrier


Goblet cells
Produce mucins that are essential for the formation of a thick mucous
layer, which covers the surface of the gut epithelium and provides
protection

Paneth cells

Secret antimicrobial peptides that break down bacterial cell membrane



Some antimicrobial peptides are expressed constitutively (i.e. α- and βdefensins, CRS peptides, and lysozyme)
Many antimicrobial peptides are induced in response to invading
pathogens (i.e. cathelicidins, and angiogenin 4)
Other epithelial cells can produce antimicrobial peptides as well
Initial Encounter
Pattern Recognition Receptors (PRRs) detects pathogen-associated molecular
patterns (PAMPs). The first PRRs to detect the presence of Salmonella via
PAMPs are the following Toll-like receptors (TLRs)…

TLRs present on the outer membrane of the cell

TLR1, 2, 4, 5, 6, 10

TLRs present in intracellular vesicles

TLR3, 7, 8, 9, 11, 13

What PAMPs are been detected?




LPS (TLR4)
Bacterial lipoproteins (TLR1/2/6)
Flagellin FliC (TLR5) (NLRC4)
Bacterial DNA (TLR9), RNA, and more
TLR Signaling Process
Ligand binding between TLRs and PAMPs
TLRs interact with adaptors MyD88 and TRIF
Induction of signaling cascades
Activation of transcriptional factors NFkB
and IRF3
Production of inflammatory cytokines (next
slide) followed by a type I IFN response
Detection by TLRs
TLRs detect extracellular Salmonella





TLR signaling
Induces increased secretion of
antimicrobial peptides RegIIIβand
RegIIIγ, which aids in the clearance of
intestinal pathogens
Mice lacking MyD88 fails to produce
RegIIIγ
Up-regulates expression of proinflammatory cytokines (i.e. IL-23)
Promotes the production of IL-8, IL10, pro-IL1β, pro-IL18, and more
Broz, P., et. al,
2012
NLR Signaling Process
Ligand binding between NLRs and PAMPs
Ligand binding between NLRs and PAMPs
Induction of various signaling
cascades
Induction of the assembly of
inflammasome, a signaling
complex
NOD1 and NOD2 interact with
RIP2 kinase
Adaptor protein ASC brings proCaspase-1 to inflammasome
RIP2 kinase activates
transcriptional factors NFkB
Activation of pro-Caspase-1 via
dimerization and autoproteolytic
cleavage
Production of inflammatory
cytokines
Secretion of mature cytokines via
activated protease-mediated
cleaving of IL-1β and IL-18
Detection by NLRs
NLRs detect intracellular Salmonella



NOD-like Receptors
Mice lacking NOD1/2 or RIP2 showed
decreased levels in inflammatory
cytokines and increased colonization in
mucosa
NLRC4/NLPR3 inflammasomes detect
T3SS (virulence factor) and activate
Caspase-1, inducing pyroptosis, a proinflammatory cell-death program
Unlike apoptosis, pyroptosis intensifies the
inflammatory response via subsequent
release of pro-inflammatory cytokines and
cellular contents, once again making
pathogens vulnerable towards extracellular
immune defenses.
Broz, P., et. al,
2012
Inflammation of Gut Mucosa
Secretion of cytokines induced by PRRs






IL-18 and IL-23
Induce the inflammatory response via
paracrine signaling mechanisms
T cells release increased amount of
IFNγ under the influence of IL-18
IL-23 induces the release of IL-22 and
IL-17, leading to increased production
of mucins and antimicrobial peptides
Innate Lymphoid Cells (i.e. NK cells)
may also produce IL-22
IL-17 promotes release of CXC
chemokines, resulting in an influx of
neutrophils into the mucosa
Broz, P., et. al,
Recruitment of Neutrophils
As an intracellular pathogen, Salmonellae
can be found extracellularly after
transcytosis or host cell lysis (pyroptosis).





Infiltrating neutrophils
IL-17-/- mice demonstrate reduced
amount of neutrophils recruited to the
intestinal mucosa
Caspase-1 deficient mice, lacking
secretion of IL-1β, produce less CXC
chemokines than wild-type mice
Play an important role in the elimination
of extracellular Salmonellae in the gut
Promote diarrhea and inflammation due
to intestinal tissue damage
Broz, P., et. al,
Macrophages & Dendritic Cells
Once crossed the epithelial cell barrier, Salmonella face other immune
defences in Gut-Associated Lymphoid Tissue GALT…

Macrophages and Dendritic Cells (DCs)

Eliminate pathogens via phagocytosis


Activate other immune cells, mainly the adaptive immune system, directly or via
producing pro-inflammatory cytokines
Effector SipB in macrophages can activate Caspase 1, producing IL-1β and IL18
Modified from LaRock, D. L., et. al, 2015
Antigen-Presenting Cells (APCs)
During Salmonella infection, professional APCs – Macrophages and
DCs…

Crucial to the activation of T cells

Process and present antigen (Ag) to T cells
Upregulate the expression of the following molecules

Costimulatory surface molecules: CD40, CD80, and CD86

Cytokines: IFNγ, TNF-α, IL-12, and IL-18

Dendritic Cells


Essential activators of naïve T cells with
diverse subpopulations
Present in the subepithelial area of PP

Where initial encounter with pathogen takes place
The primary APCs during early stages of infection

Express low levels of lysosomal proteases





Take longer than microphages to degrade bacterial
proteins
Environment surrounding mature DC is more
supportive of Ag processing and presentation than
elimination of bacteria
Stronger induction of IFN-γ production by
CD4+ T cells
Contribute more to Th1-type responses than
macrophages
Immature Ag-capturing
cells (Phagocytic)
Undergo morphological
and functional changes
after bacterial uptake
Mature APCs
Macrophages

Research suggests macrophages contribute more to the
production of proinflammatory chemokines and cytokines
than DCs

Chemokines: CCl5, CCL20, CXCL10

Cytokines: IL-18, TNF-α



Macrophage-depleted mice can generate CD8+ T cells
against Salmonella-specific antigens
More essential in inducing proinflammatory responses rather than
carrying out APC function
May be more efficient in the stimulation of effector T cells rather than
naïve T cells
Natural Killer (NK) Cells

NK cells are activated by macrophages, leading to

Secretion of IFN-γ

Degranulation

Decreased number of live bacteria inside macrophages

Interaction between NK cells and infected macrophages

Contact-dependent

IL-2 and/or IL-15 required to prime NK cells to produce IFN-γ

Infected macrophages secrete IL-12 and IL-18, which activate NK cells
Lapaque, N., et. al, 2009
Types of T Lymphocytes

Express heterodimeric receptors (TCRs) on surface

TCRs interact with Ag

Most of TCRs consist of one α- and one β- chain

One group expresses CD4 coreceptors
One group expresses CD8 coreceptors

A minority of TCRs consist of one γ- and δ- chain






Abundant in epithelial tissues, especially in the gut mucosa
Less diverse than αβT cells
MHC presentation of Ags is not required for activation
May have a role in both innate and adaptive immune responses
Both αβ T cells and γδ T cells are detected after Salmonella
infections; however the role of γδ T cells is not well understood.
Wolfert, M. A. and Boons, G., 2013
Modified from Wolfert, M. A. and Boons, G., 2013
Activation of CD4+ T Cell
TCRs interact with bacterial peptides presented on MHC-II molecules



Peptides obtained from endocytic or phagocytic vacuoles
CD4+ T cells recognize FliC, an abundant subunit protein found in the flagella
Salmonella invasion protein SipC is also an immune target
CD4+ T Cells

Express CD4 co-receptors on surface

Responsible for production of cytokines

Induce B-cell class switching and affinity maturation

Stimulate activation and growth of functional cytotoxic T cells

Mediates macrophage activation
IFN-γ Production



Essential for clearance of infection
CD4+ T-cell-deficient mice produce 100-fold less IFN-γ than wild-type mice,
failing to clear an infection with an attenuated strain
Reduced IFN-γ or CD4+ T cells lead to reactivation of latent Salmonella
infection
Wolfert, M. A. and Boons, G., 2013
Activation of CD8+ T Cell
TCRs interact with endogenous antigens presented on MHC-I molecules



CD8+ cells are activated, becoming cytotoxic T cells
Recognize peptides derived from GroEL
More research is required to identify additional Ags targeted by T cells
CD8+ T Cells

Express CD8 co-receptors on surface

Involved in secretion of cytokines

Involved in bacterial clearance

Mice lacking MHC-I have higher bacterial loads during primary infection than
wild-type mice

TCR-deficient mice develop more severe infection than MHC-II knockouts

Re-expose intracellular pathogen to phagocytes
Granulysin


A protein found in cytotoxic T cell (CTL) granules
CD4+ T-cell-deficient mice produce 100-fold less IFN-y than wild-type mice,
failing to clear an
Modified from Wolfert, M. A. and Boons, G., 2013
Activation of B Cells
B cells that present the same antigenic peptides on MHC-II molecules as the
ones on activated TH cells can become activated via cytokines.

Antibody-secreting plasma cells

Memory B Cells
B Cells

Antibody production

Rely on help from T-cell for antibody isotype switching

O-antigen polysaccharide chains on LPS are the main targets

Also generated against unknown antigenic proteins

Mediate the priming of T cells and the generation of memory T-cells


Influence immune response based on the cytokines produced (IFN-y: Th1-type
response; IL-4: Th2- type response)
Activated B cells function as APCs to prime naïve T cells (important in the spleen)
Secretory IgA (sIgA)



Present in mucosal tissues
Reduces adherence of pathogen to cells, but not required for protective
immunity
Mice that cannot generate Salmonella-specific sIgA are able to mount a same
protective immune response as wild-type mice in Typhimurium infection
General Overview of Intestinal Immunity
General Overview of Intestinal Immunity


Salmonella in the gut lumen encounter initial barriers (i.e.
antimicrobial peptides and sIgA) and cross the epithelium via M cells
Bacteria express FliC protein, which is recognized by TLR5, initiating
an inflammatory response along with production of cytokines

Cytokines lead to an influx of macrophages, neutrophils, and DCs

DC or Macrophage-mediated phagocytosis of bacteria  Pyroptosis



Bacteria persist within phagocyte after its uptake  Inhibited T-cell activation
and increased expression of MHC and co-stimulatory molecules on DCs
Degradation of bacteria within neutrophils or macrophages  DCs in the PP
present processed Ags to naïve T cells and enhance activation of T cells via
cytokine production (TNF-α and IL-12)
Activated TH cells produce cytokines that activate B cells, which
proliferate into antibody-secreting plasma cells and memory B cells
Q2: Host Damage
What damage ensues to the host from the
immune response?
The Microbiota
Gut Microbiota Functions
 Required for the proper development of immune system
 Has important homeostatic immune & metabolic functions
 Affects the proliferation and survival of epithelial cells
 Provides protection against pathogens
Dysbiosis


Inflammation from host immune response may result in disequilibria
between the host and microbiota, aka dysbiosis
Increased risk for neoplatic transformation in host
Zitvogel et al., 2015
Inflammation and Colorectal Cancer

Inflammation contributes to tumour development

Premalignant cells may activate proliferation and anti-apoptotic mechanisms

Activated inflammatory cells produce ROS, which promotes DNA damage and
mutation
Teric et al., 2010
Tumour-Promoting
Cytokines


Teric et al., 2010
Intestinal
inflammation
caused by microbial
pathogens can lead
to tissue injury,
which may promote
tumour if
inflammation
becomes chronic
Inflammation may
also lead to septic
shock, organ failure,
and/or chronic
inflammatory
diseases
Q3: Bacterial Evasion
How do the bacteria attempt to evade these
host response elements.
Overview of
Immune Evasion
 Pyroptosis of
APC



Cummings, L. A. et. al, 2009
Inhibition of Ag
Processing and
Presentation
Inhibition of T
Cell Activation
Intracellular
Replication
Caspase-1-Induced Pyroptosis
From death comes life…
 Evasion of NLRC4 (Reminder: NLRC4
recognizes FliC and T3SS)





SPI2 T3SS rod protein, SsaI, changes amino
acid to avoid being detected by NLRC4
Presence of SPI2 T3SS results in suppressed
expression of flagellin
NLRC4 can still detect pathogen, however at a
delayed time post-infection
With expression of FliC, NLRC4 is triggered
within 6h; 17h without FliC
Leads to intracellular replication of bacteria in
macrophages via suppression of pyroptosis
Miao, E.A. and Rajan, J.V., 2011
Antigen Processing & Presentation




Salmonella alter DC functions, compromising activation of T cells
SPI-2 genes encode virulence proteins (i.e. SpiC) that are secreted into the
cytoplasm to prevent Ag processing by avoiding phagosome-lysosome fusion
These virulence proteins target P13K activity, preventing bacterial degradation
Salmonella can reduce expression of bacterial antigens such as flagellin and LPS
in MLNs and spleen, preventing their presentation to T cells
Bueno, S.M. et. al, 2007
Intracellular Survival






Avoid neutrophil-mediated killing by invading, surviving, and
replicating inside epithelial cells, macrophages, and DCs
Salmonellae rely on bacterial membrane remodeling via regulatory
proteins to enhance intracellular survival
Spi-2 type III secretion system secrete effectors such as SifA to change the
composition of the Salmonella-containing vacuole, preventing phagosome
acidification (avoid NO- and NADPH oxidase-mediated killing)
Modify lipid A and other components of LPS via mechanisms such as
deacylation and palmitylation to reduce recognition by TLR2 and 4
PgtE, an outer membrane expressed on Salmonella, cleaves αantimicrobial peptides, promoting its resistance to innate immune response
While inside phagocytes, Salmonella can spread to other parts of
the body (i.e. liver and spleen) for colonization
Q4: Outcome
Is the bacteria completely removed, does the
patient recover fully and is there immunity to
future infections with these candidate infectious
agents?
Salmonella persistence



Most people are able to have the bacteria completely cleared, but
some become carriers of Salmonella
Salmonella infections can survive activation of the innate and
adaptive immune responses, and remain in the host
“immune status-quo” is reached, meaning an equilibrium is established
between the immune system and bacteria

Mostly found in the mesenteric lymph nodes, sometimes in the spleen and liver

During persistent infections, pathogens ensure survival by

Undergoing antigenic variation and antigenic imitation

Inhibition synthesis of host proteins

Inactivation of humoural immune components

Avoiding immune recognition by hiding
Immune System in Chronic Infections







Macrophages
Research suggests that Salmonella becomes dormant after entering
macrophages
~80% of the bacteria are found within MOMA-2 expressing macrophages in
mesenteric lymph nodes (MLN)
Dendritic Cells
Can transport Salmonella from the GI tract to MLN; transportation is mainly
accomplished by CD103+ CD11B+ DCs
Decreased intracellular proliferation of Salmonella within DC affects antigen
presentation, reducing T-cell responses, allowing for persistent infection
A subset of DCs carrying pathogen can migrate from the lamina propria into
the intestinal lumen, leading to shedding of live bacteria in stools (transmission)
Adaptive Immunity in Chronic Infections

Early resistance phase (acute infection)

High Th1 (pro-inflammatory) and low Th2 (anti-inflammatory) responses

Maintenance of “Immune Status-quo”


A lower Th1 response; however production IFN-y is still required as reactivation
of acute infection is observed in mice injected with anti-IFN-y antibodies
Regulatory T cells suppress Th1 response
Microbiota


Needed to reduce bacterial load in the gut and stools during chronic
infections
Infected individuals may shed bacteria from a year up to lifetime
Protection Against Future Infections





Strong, persistent B- and T-cell memory is
established
IFN-y-secreting memory T-Cell populations are
detected in vaccinated subjects
Memory B cells are crucial in fighting future infections
as well because humoural protection is positively
correlated with CD4+ T cell IFN-y production
Circulating antibodies also help protect against future
infections
B cells are crucial in the development of Th1 memory
cells
References – Q1 & Q4






Broz, P., Ohlson, M. B., & Monack, D. M. (2012). Innate immune response to
salmonella typhimurium, a model enteric pathogen. Gut Microbes, 3(2), 62-70.
Bueno, S. M., Riquelme, S., Riedel, C. A., & Kalergis, A. M. (2012). Mechanisms
used by virulent salmonella to impair dendritic cell function and evade
adaptive immunity. Immunology, 137(1), 28-36.
Cummings, L. A., Deatherage, B. L., & Cookson, B. T. (2009). Adaptive immune
responses during salmonella infection. EcoSal Plus, 3(2),
10.1128/ecosalplus.8.8.11.
Griffin, A. J., & McSorley, S. J. (2011). Development of protective immunity to
salmonella, a mucosal pathogen with a systemic agenda. Mucosal
Immunology, 4(4), 371-382.
Kupz, A., Scott, T. A., Belz, G. T., Andrews, D. M., Greyer, M., Lew, A. M., et al.
(2013). Contribution of Thy1+ NK cells to protective IFN-gamma production
during salmonella typhimurium infections. Proceedings of the National Academy
of Sciences of the United States of America, 110(6), 2252-2257.
Lapaque, N., Walzer, T., Meresse, S., Vivier, E., & Trowsdale, J. (2009).
Interactions between human NK cells and macrophages in response to
salmonella infection. Journal of Immunology (Baltimore, Md.: 1950),182(7),
4339-4348.
References – Q1 & Q4







LaRock, D. L., Chaudhary, A., & Miller, S. I. (2015). Salmonellae interactions
with host processes. Nature Reviews.Microbiology, 13(4), 191-205.
Mittrucker, H. W., & Kaufmann, S. H. (2000). Immune response to infection
with salmonella typhimurium in mice. Journal of Leukocyte Biology, 67(4),
457-463.
Perez-Lopez, A., Behnsen, J., Nuccio, S. P., & Raffatellu, M. (2016). Mucosal
immunity to pathogenic intestinal bacteria. Nature Reviews.Immunology, 16(3),
135-148.
Pham, O. H., & McSorley, S. J. (2015). Protective host immune responses to
salmonella infection. Future Microbiology, 10(1), 101-110.
Wolfert, M. A., & Boons, G. J. (2013). Adaptive immune activation:
Glycosylation does matter. Nature Chemical Biology, 9(12), 776-784.
Xavier, R. J., & Podolsky, D. K. (2007). Unravelling the pathogenesis of
inflammatory bowel disease. Nature, 448(7152), 427-434.
Xavier, R. J., & Podolsky, D. K. (2007). Unravelling the pathogenesis of
inflammatory bowel disease. Nature, 448(7152), 427-434.
References – Q2






Rakoff-Nahoum, S., and R. Medzhitov. "Role of toll-like receptors in tissue repair and
tumorigenesis." Biochemistry (Moscow) 73.5 (2008): 555-561.
Uronis, Joshua M., et al. "Modulation of the intestinal microbiota alters colitisassociated colorectal cancer susceptibility." PloS one 4.6 (2009): e6026.
Terzić, Janoš, et al. "Inflammation and colon cancer." Gastroenterology 138.6 (2010):
2101-2114.
Plottel, Claudia S., and Martin J. Blaser. "Microbiome and malignancy." Cell host &
microbe 10.4 (2011): 324-335.
Huber, Samuel, et al. "IL-22BP is regulated by the inflammasome and modulates
tumorigenesis in the intestine." Nature 491.7423 (2012): 259-263.
Zitvogel, Laurence, et al. "Cancer and the gut microbiota: An unexpected link."Science
Translational Medicine 7.271 (2015): 271ps1-271ps1.
References – Q3








Bueno, S. M., Gonzalez, P. A., Carreno, L. J., Tobar, J. A., Mora, G. C., Pereda, C. J., et
al. (2008). The capacity of salmonella to survive inside dendritic cells and prevent
antigen presentation to T cells is host specific. Immunology, 124(4), 522-533.
Bueno, S. M., Gonzalez, P. A., Schwebach, J. R., & Kalergis, A. M. (2007). T cell immunity
evasion by virulent salmonella enterica. Immunology Letters, 111(1), 14-20.
Cummings, L. A., Deatherage, B. L., & Cookson, B. T. (2009). Adaptive immune responses
during salmonella infection. EcoSal Plus, 3(2), 10.1128/ecosalplus.8.8.11.
Finlay, B. B., & McFadden, G. (2006). Anti-immunology: Evasion of the host immune
system by bacterial and viral pathogens. Cell, 124(4), 767-782.
Finlay, B. B., & McFadden, G. (2006). Anti-immunology: Evasion of the host immune
system by bacterial and viral pathogens. Cell, 124(4), 767-782.
Kaur, J., & Jain, S. K. (2012). Role of antigens and virulence factors of salmonella
enterica serovar typhi in its pathogenesis. Microbiological Research, 167(4), 199-210.
Miao, E. A., & Rajan, J. V. (2011). Salmonella and caspase-1: A complex interplay of
detection and evasion. Frontiers in Microbiology, 2, 85.
Purushothaman, S. S., Wang, B., & Cleary, P. P. (2003). M1 protein triggers a
phosphoinositide cascade for group A streptococcus invasion of epithelial cells. Infection
and Immunity, 71(10), 5823-5830.