immunity responds to listeria monocytogenes

65
1

Upload: moein-ybeygi

Post on 08-Jul-2015

235 views

Category:

Education


0 download

DESCRIPTION

These Slides are my presentation in M.s degree in Immunology lesson for my Classmates at my university. You can found important information about Virulence factors of Listeria monocytogenes that can Scape from immune responds and defeat them.

TRANSCRIPT

Page 1: Immunity Responds to Listeria monocytogenes

1

Page 2: Immunity Responds to Listeria monocytogenes

Qazvin university of

Medical Science, Fall 1392

2

Page 3: Immunity Responds to Listeria monocytogenes

Listeria monocytogenes is a Gram-positive pathogenic bacterium that has adapted to various environments, from soils and food products to the intestinal tract and intracellular compartments of diverse animal species and humans.

The aerobic

Non–spore-forming

Catalse Positive

The organisms are motile at room temperature (25 ̊C) but less so at 37 °C

L. monocytogenes exhibits weak β-hemolysis when grown on sheep blood agar plates.

3

Page 4: Immunity Responds to Listeria monocytogenes

L. monocytogenes is a facultative intracellular pathogen

that can live both inside and outside its host.

To infect its mammalian host and to cause the most severe

pathologies, L. monocytogenes is able to cross :

The intestinal

Blood-brain and

Maternofetal barriers

Crossing the host barriers involves bacterial invasion and

survival within a large variety of normally nonphagocytic

cells

It is therefore crucial to understand how L. monocytogenes

induces its own uptake by host cells.

4

Page 5: Immunity Responds to Listeria monocytogenes

Listeriosis manifests itself through flu-like symptoms

and can lead to

diarrhea,

meningitis,

encephalitis,

meningoencephalitis and

Stillbirths

In humans, it primarily infects immunocompromised

individuals like

pregnant women,

neonates, and

the elderly

5

Page 6: Immunity Responds to Listeria monocytogenes

6

Page 7: Immunity Responds to Listeria monocytogenes

Listeriosis is a severe foodborne disease characterized by bacteremia and meningoencephalitis in individuals with impaired cell-mediated immunity, Including :

Neonates,

Pregnant woman,

Elderly persons, and

Immunosuppressed patients.

The potential of L. monocytogenes to cause disease correlates with its capacity to survive within macrophages, to invade nonphagocytic cells and replicate therein and also to cross the intestinal, the blood–brain, and the fetoplacental barriers.

7

Page 8: Immunity Responds to Listeria monocytogenes

The incidence of listeriosis is rather low, compared to

other common foodborne pathogens such as

Campylobacter species, Salmonella species, Shigella

species, andVibrio species.

However

The outcome is much more severe and often fatal.

In fact, it represents one of the most deadly bacterial

infections due to its high mean mortality rate of 20%–

30%, despite early antibiotic treatment.

8

Page 9: Immunity Responds to Listeria monocytogenes

9

Page 10: Immunity Responds to Listeria monocytogenes

Internalins Proteins (ex : InL A, InL B ,....)

Listeriolysin O (LLO)

Phospholipases

Act A

The Virulence Genes (prfA, PlcA, hly, mpl,...)

PI-PLC

PC-PLC

10

Page 11: Immunity Responds to Listeria monocytogenes

There exist two principle mechanisms by which L.

monocytogenes can enter into the host through the

intestinal mucosa.

The first route is direct invasion of the enterocytes

lining the absorptive epithelium of the

microvilli, leading to infection of the intestinal cells

This entry mechanism occurs only in humans and some

susceptible animals.

The second entry pathway is translocation across the

M-cells of Peyer’s patches.

11

Page 12: Immunity Responds to Listeria monocytogenes

Internalin (InlA), and InlB were the first surface proteins of L.monocytogenes identified to promote host cell invasion.

They have an amino-terminal leucine-rich repeat domain(LRR) formed by tandem repeats of 20-22 amino acids.

The LRR regions are structurally and functionally important to theninternalization of LM.

LRRs provide versatile recognition units for protein-protein interactions and protein activation in a variety of prokaryotic and eukaryotic proteins.

12

Page 13: Immunity Responds to Listeria monocytogenes

L. monocytogenes genome encodes 22 additional proteins

containing LRRs that form the internalin family.

The amino-terminal LRRs are followed by a conserved

inter-repeat domain (IR)

The carboxy-terminal LPXTG motif, which mediates

covalent binding to the cell wall peptidoglycan, is present

in 19 members including the internalin prototype, InlA

One member, InlB, is bound to the lipoteichoic acids on

the bacterial cell wall by electrostatic interactions

involving carboxy-terminal glycine/tryptophan-rich

(GW) modules.

13

Page 14: Immunity Responds to Listeria monocytogenes

14

Page 15: Immunity Responds to Listeria monocytogenes

15

Page 16: Immunity Responds to Listeria monocytogenes

InlA, an 800-amino-acid protein, is responsible for

bacterial entry into epithelial cells.

InlA specifically interacts with E-cadherin.

E-cadherin: a member of the cadherin superfamily of

calcium-dependent cell adhesion molecules.

E-cadherin is involved in the formation of adherens

junctions in polarized epithelial cells of different tissues.

16

Page 17: Immunity Responds to Listeria monocytogenes

17

Page 18: Immunity Responds to Listeria monocytogenes

The carboxyl terminal of E-cadherin directly interacts

with the intracellular β-catenin.

α-catenin, in turn, binds to β-catenin and interacts with

actin This interaction leads to the formation of a fusion

molecule consisting of the ectodomains of the E-

cadherin and the actin binding site of the α-catenin

which eventually leads to LM entry.

Furthermore, myosin VIIA and its ligand vezatin

together function as the molecular motor in the

internalization of Listeria.

When myosin VIIA binds vezatin, coupled with an actin

polymerization process, it provides the tension

necessary for bacterial internalization.

18

Page 19: Immunity Responds to Listeria monocytogenes

19

Page 20: Immunity Responds to Listeria monocytogenes

InlB, a 630-amino-acid protein, promotes

L. monocytogenes entry into a large variety of mammalian cells including :

Epithelial cells

Endothelial cells

Hepatocytes

Fibroblasts

The hepatocyte growth factor receptor (Met/HGF-R) has

been identified as the major ligand for InlB responsible for

L. monocytogenes entry into non-phagocytic cells, and for

cell scattering and membrane ruffling induced by soluble

InlB

20

Page 21: Immunity Responds to Listeria monocytogenes

Met belongs to the family of receptor tyrosine kinases

(RTKs), one of the largest and most important families of

transmembrane signaling receptors expressed by a large

variety of cells.

Met plays crucial roles in :

Organ morphogenesis,

Cell proliferation,

Cell migration and differentiation,

And also in cell growth and invasion during metastasis

in cancer cells

21

Page 22: Immunity Responds to Listeria monocytogenes

InlB functionally mimics HGF, the natural Met

ligand, through the binding of its LRRs.

As in the case of the InlA/E-cadherin

interaction, InlB interaction with Met is species

specific: InlB interacts with human and mouse

Met, but does not recognize the guinea-pig or rabbit

receptor.

22

Page 23: Immunity Responds to Listeria monocytogenes

23

Page 24: Immunity Responds to Listeria monocytogenes

24

Page 25: Immunity Responds to Listeria monocytogenes

Listeriolysin O (LLO), the major and first identified

virulence factor of L. monocytogenes is a member of the

cholesterol-dependent cytolysin (CDC) family of toxins.

These toxins are produced by numerous Gram-positive

bacterial pathogens including :

Streptococcus pneumonia

Bacillus anthracis.

Among these pathogens only L. monocytogenes is known

to infect nonphagocytic cells and LLO, secreted by L.

monocytogenes, is required for bacterial escape from

endocytic vacuoles.

25

Page 26: Immunity Responds to Listeria monocytogenes

LLO binds to the host plasma membrane as a monomer

and then forms oligomers composed of up to 50 subunits

that inserts into the plasma membrane forming pores of

about 200e 300 A° diameter.

hly, one of the many genes activated during

infection, leads to the production of Listeriolysin O

(LLO).

Unlike the other CDCs, pore formation by LLO is more

efficient at low pH facilitating the disruption of endocytic

membranes following acidification of the vacuoles.

This is a crucial function for cellular invasion of both

phagocytic and nonphagocytic cells.

26

Page 27: Immunity Responds to Listeria monocytogenes

LM also uses phospholipases C to aid in the escape from the vacuole.

Two specific phospholipases (PLCS) are used.

One is the phosphatidylinostiol-specific PLC (PI-PLC), and

The other is more general, phosphatidylinostiol-specific (PC-PLC).

The role of the PI-PLC secreted by LM is to catalyze the production of inositol phosphate and diacylglycerol (DAG) through cleavage of the membrane lipid PI.

DAG then has the ability to activate protein kinase C (PKC).

There are four types of PKCs, but the PKC of the host is shown to be linked with the PI-PLC signaling cascade.

The PKC has been shown to facilitate the permeation of the phagosomal membrane before the bacteria escape.

27

Page 28: Immunity Responds to Listeria monocytogenes

In summary, the secretion of PLCs during listerial

infection has several effects on the host cell.

One of these effects has been shown to :

1) Increase the permeation of phagosomal membrane.

2) The activation of PKC through PI-PLC facilitates the

escape of the bacterium.

3) The decreased affinity of L. monocytogenes for the

glycan linker of the GPI-anchored protein due to the

lack or absence of the Vb-strand also increases the

ability for the bacterium to escape during infection.

4) PC-PLC leads to the activation of NF-kB, which allows

the bacteria to exploit the host cell machinery.

28

Page 29: Immunity Responds to Listeria monocytogenes

The bacterial surface protein ActA is a major virulence

factor of L. monocytogenes that enables bacterial

propulsion in the cytosol leading to the invasion of yet

uninfected neighboring cells by a process called cell-to-

cell spreading.

Host cell interaction could be mediated by the amino-

terminal region of ActA that has several clusters of

positively charged amino-acids that could bind heparan

sulfate proteoglycans.

The precise mechanism involved in ActA -mediated

invasion remains to be elucidated.

29

Page 30: Immunity Responds to Listeria monocytogenes

30

Page 31: Immunity Responds to Listeria monocytogenes

31

Page 32: Immunity Responds to Listeria monocytogenes

32

Page 33: Immunity Responds to Listeria monocytogenes

33

Page 34: Immunity Responds to Listeria monocytogenes

34

Page 35: Immunity Responds to Listeria monocytogenes

35

Page 36: Immunity Responds to Listeria monocytogenes

After infection within the gastrointestinal tract

immediate immune responses are essential for the

control of pathogens, such as L. monocytogenes.

Activation of the innate immune system is triggered

when pathogen-associated molecular patterns (PAMPs)

engage pattern recognition receptors (PRRs) on

intestinal epithelial cells (IECs).

36

Page 37: Immunity Responds to Listeria monocytogenes

Typical PAMPs include bacterial carbohydrates, such as

Lipopolysaccharide (LPS)

Mannose

Nucleic acids (both DNA and RNA)

Peptidoglycan components

Lipoteichoic acids, and probably many other

molecules, and are able to trigger the innate immune

response.

37

Page 38: Immunity Responds to Listeria monocytogenes

Innate immunity to L. monocytogenes is primarily

mediated by two types of pattern recognition receptors:

The Toll-like receptors (TLRs)

The nucleotide-binding oligomerization domain

(NOD)-like receptors (NLRs).

In addition, there is some experimental evidence for

the involvement of scavenger receptors and a TLR-9

independent cytosolic sensor system for bacterial DNA

38

Page 39: Immunity Responds to Listeria monocytogenes

Upon recognition of the presence of microbes

through sensing pathogen-associated molecular

patterns.

TLRs can bind any of the 4 known activating

adaptors:

Myeloid differentiating factor-88 (MyD88)

MyD88 adapter-like (Mal)

TIR domain-containing adapterinducing IFN-ɣ(TRIF)

TRIF-related adapter molecule (TRAM)

39

Page 40: Immunity Responds to Listeria monocytogenes

MyD88 appears to be the key adaptor

molecule, because it is required for signaling by all

TLRs with only one exception: TLR3 uses TRIF.

The binding of the activating adaptors results in the

subsequent recruitment of IL-1R, associated kinases

(IRAKs) and downstream activation of transcription

factors including NF-κB and IFN regulatory factor 3

(IRF3), which in turn induces the proinflammatory

cytokines and type I IFNs.

40

Page 41: Immunity Responds to Listeria monocytogenes

Toll-like receptor 2 (TLR2) can interact with several

specific ligands, including :

Bacterial lipoproteins

Lipoteichoic

Acids of Gram-positive bacteria such as L.

monocytogenes

Yeast zymosan

TLR2 can form heterodimers with TLR1 and

TLR6, thereby improving the recognition of the target

lipoteichoic acids.

41

Page 42: Immunity Responds to Listeria monocytogenes

TLR2 is expressed on the cell surface of intestinal

epithelial cells and its activation by commensal

bacteria is thought to play an important role in the

maintenance of the integrity of the intestinal epithelial

barrier.

TLR2 is also expressed within

phagolysosomes, thus, L. monocytogenes cells that

have escaped into the host cell cytoplasm were not

detected by TLR2.

The importance of TLR2 signaling for early protection

against L. monocytogenes is, however, inconclusive.

42

Page 43: Immunity Responds to Listeria monocytogenes

Toll-like receptor 5 (TLR5) can bind to a protein motif

common to the flagellin protein making up the flagella

from many bacteria, such as L. monocytogenes.

TLR5 activation induces NF-κB and stimulates TNF

production, suggesting that TLR5 may serve as a

general alarm system, when the gastrointestinal barrier

is compromised by a broad spectrum of motile bacteria.

43

Page 44: Immunity Responds to Listeria monocytogenes

Toll-like receptor 9 (TLR9) recognizes the CpG motifs

present in bacterial DNA. In immune cells, TLR9 is

exclusively localized in the endosomes.

In the intestine, TLR9 was shown to be located on

both, the apical and the basolateral surface of IECs.

Upon activation of TLR9, IκBα is degraded and NF-κB

is activated, again resulting in a proinflammatory

response.

44

Page 45: Immunity Responds to Listeria monocytogenes

In conclusion, the available experimental evidence

suggests that TLR2 is the most relevant TLR for

recognition of L. monocytogenes cells However, as

IECs show.

TLR2 commensal ligand-induced activation, TLR2 is

also considered to play an important role in maintaining

the integrity of the intestinal epithelial barrier.

45

Page 46: Immunity Responds to Listeria monocytogenes

46

Page 47: Immunity Responds to Listeria monocytogenes

47

Page 48: Immunity Responds to Listeria monocytogenes

48

Page 49: Immunity Responds to Listeria monocytogenes

The NLRs are critical for mucosal innate immunity as

sensors of microbial components and cell injury in the

cytoplasm.

Both NOD1 and NOD2 are important for the innate

immune response against L. monocytogenes, because

they represent intracellular sensors of bacterial

peptidoglycan components that are thought to enter

cells by endocytosis through clathrin-coated pits.

49

Page 50: Immunity Responds to Listeria monocytogenes

NOD1 is ubiquitously expressed in adult human

Tissues.

NOD2 is expressed only in leukocytes, DCs, and

epithelial cells.

Activation of NOD1 and NOD2 results in

the translocation of NF-κB and mitogen-activated protein

kinase into the nucleus, to up-regulate the transcription of

proinflammatory genes and mediate antibacterial effects

by the up-regulation of another group of small

antibacterial peptides, the defensins.

50

Page 51: Immunity Responds to Listeria monocytogenes

NOD1 recognizes a diaminopimelic acid-containing

dipeptide or tripeptide molecule generated by lysozyme

action on the peptidoglycan of many Gram-negative and

Gram-positive bacteria, including L. monocytogenes.

NOD2 is activated by muramyl dipeptide

(MDP), which is another degradation product of the

peptidoglycan produced by lysozyme and other

(bacterial) peptidoglycan hydrolases.

51

Page 52: Immunity Responds to Listeria monocytogenes

52

Page 53: Immunity Responds to Listeria monocytogenes

DCs respond to different pathogens and initiate the appropriate type of T cell response needed to control the infection.

In response to L. monocytogenes infection, DCs are critical in priming the T cell response, since mice depleted of DCs are unable to generate a CD8 T cell response.

Due to the primarily intracellular localization of L. monocytogenes, CD4 and CD8 T cells mediate most of the adaptive immune response, and are crucial for long-termimmunity after initial L. monocytogenes infection.

53

Page 54: Immunity Responds to Listeria monocytogenes

Almost any cell type that harbors L. monocytogenes in the cytoplasm can process the proteins secreted from the pathogen, by degradation and subsequent loading on MHC class I molecules, in order to present them on the cell surface to CD8 T cells.

Only professional antigen presenting cells (APCs) can present antigens derived from lysosomal degradation via the MHC class II pathway to CD4 T cells.

54

Page 55: Immunity Responds to Listeria monocytogenes

The CD8 T cells mediate the anti-Listeria immunity by

two synergistic mechanisms:

first:

by secretion of IFN-γ to activate macrophages;

Secondly:

by lysis of infected cells via perforin and

granzymes, leading to the exposure of intracellular

bacteria to the activated macrophages

55

Page 56: Immunity Responds to Listeria monocytogenes

IFN-γ is known to be essential for host resistance to

intracellular pathogens such as L. monocytogenes, as it

mediates the activation of resting macrophages that

more efficiently restricts the multiplication of

intracellular pathogens and promotes long-term

protective cellular immunity.

56

Page 57: Immunity Responds to Listeria monocytogenes

L. monocytogenes induces a strong T-helper type 1

response and, similar to CD8 T cells, CD4 T cells

also secrete IFN-γ.

The strong CD8 and CD4 T cell responses results in a

stable population of memory T cells specific for L.

monocytogenes

57

Page 58: Immunity Responds to Listeria monocytogenes

In the intestine, NKT cells (lymphocytes expressing

both NK and T cell markers) play an important role in

the control of early infection with L. monocytogenes.

In general, adaptive immune responses in the intestine

are characterized by high numbers of IgA producing

plasma cells, regulatory T cells, and IL-17 producing T

cells whose development is closely linked to factors

produced by PRRs expressing IECs, DCs, and

macrophages.

58

Page 59: Immunity Responds to Listeria monocytogenes

59

Page 60: Immunity Responds to Listeria monocytogenes

60

Page 61: Immunity Responds to Listeria monocytogenes

61

Page 62: Immunity Responds to Listeria monocytogenes

62

Page 63: Immunity Responds to Listeria monocytogenes

63

Page 64: Immunity Responds to Listeria monocytogenes

64

Page 65: Immunity Responds to Listeria monocytogenes

65